ANTIGEN-BINDING MOLECULE HAVING REGULATED CONJUGATION BETWEEN HEAVY-CHAIN AND LIGHT-CHAIN

It was found that association between CH1 and CL can be suppressed by substituting amino acids that exist on the interface between CH1 and CL with electrically-charged amino acids, and that formation of heterogeneous molecules is enabled more efficiently than by introducing knobs into holes mutations into CH3 domain.

Skip to: Description  ·  Claims  · Patent History  ·  Patent History
Description
CROSS-REFERENCE TO RELATED APPLICATIONS

This application is a divisional of U.S. application Ser. No. 14/351,654, filed on Apr. 14, 2014, which is the National Stage of International Application No. PCT/JP2012/078103, filed on Oct. 31, 2012, which claims the benefit of Japanese Application Serial No. 2011-238873, filed on Oct. 31, 2011.

BACKGROUND ART

The present invention relates to antibodies with regulated association of the heavy chain and light chain, methods for producing an antibody with regulated association of the heavy chain and light chain, methods for regulating association of the heavy chain and light chain of an antibody, pharmaceutical compositions comprising that antibody as an active ingredient, and the like.

TECHNICAL FIELD

Several methods have previously been reported as methods for preparing IgG-type bispecific antibodies having human constant regions (IgG-type antibodies having a human constant region that has binding specificity for an antigen A on one arm and binding specificity for an antigen B on the other arm). In general, IgG-type bispecific antibodies are composed of two types of H chains (namely, an H chain for antigen A and an H chain for antigen B) and two types of L chains (namely, an L chain for antigen A and an L chain for antigen B). When such IgG-type bispecific antibodies are expressed, 10 types of combinations are possible as combinations of H2L2 since two types of H chains and two types of L chains are expressed. Among these, there is one type of combination that has the desired binding specificity (IgG having binding specificity for antigen A on one arm and binding specificity for antigen B on the other arm). Consequently, in order to acquire the desired bispecific antibody, it is necessary to purify one type of antibody of interest from among ten types of antibodies, which is extremely low in efficiency and difficult.

Methods have been reported for solving this problem, which involve preferentially secreting IgG having a heterologous combination of an H chain for antigen A and an H chain for antigen B, by substituting amino acids in the CH3 region of the IgG H chain (Patent Documents 1, 2, 3 and 4, and Non-Patent Documents 1 and 2). Among these, there have been reported methods that use physical obstacles in the form of a “knob” and “hole”, and those that use electric charge repulsion.

A method has also been reported for efficiently obtaining a desired molecule, which uses a common L chain in which an L chain for antigen A and an L chain for antigen B are present on a same amino acid sequence (Patent Documents 5 and 6). However, since the use of a common L chain has the potential of considerably lowering the antigen affinity, this is not necessarily the optimum method. Consequently, in order for a bispecific antibody to bind to two antigens with high affinity, it is preferable that only the L chain and H chain for antigen A associate, and only the L chain and H chain for antigen B associate. Moreover, a method has been reported to allow the H chains and L chains for each antigen to associate irrespectively of the variable regions, which comprises substituting amino acids in the CH1 and CL domains which are constant regions, instead of those in the variable regions (Patent Documents 2 and 7). However, this method is still insufficient for efficiently producing a bispecific antibody of interest.

PRIOR ART DOCUMENTS Patent Documents

  • [Patent Document 1] WO 96/27011
  • [Patent Document 2] WO 2006/106905
  • [Patent Document 3] WO 2009/089004
  • [Patent Document 4] WO 2010/129304
  • [Patent Document 5] WO 98/050431
  • [Patent Document 6] WO 2006/109592
  • [Patent Document 7] WO 2007/147901

Non-Patent Documents

  • [Non-Patent Document 1] Ridgway J B et al., Protein Engineering, 1996, Vol. 9, p. 617-621
  • [Non-Patent Document 2] Merchant A M et al., Nature Biotechnology, 1998, Vol. 16, p. 677-681

SUMMARY OF THE INVENTION Problems to be Solved by the Invention

The present invention has been achieved under such circumstances. An objective of the present invention is to provide antibodies in which association of the heavy chains and light chains is regulated, a method for producing antibodies in which the association of the heavy chains and light chains is regulated, and a method for regulating association of the heavy chains and light chains of an antibody. In addition, in one embodiment of the present invention, an objective of the present invention is to provide bispecific antibodies in which association at the interface of CH1 and CL is regulated, and a method for efficiently producing a bispecific antibody by regulating the association at the interface of CH1 and CL.

Means for Solving the Problems

The inventors of the present invention selected a constant region of the heavy chain, CH1, and a light chain constant region (CL) as heavy-chain and light-chain regions to be used for regulating association, and conducted dedicated studies on the regulation of CH1 and CL association. As a result, the present inventors found that association of CH1 and CL can be suppressed by substituting amino acid residues present at the interface of CH1 and CL with amino acid residues that mutually repel electrically or amino acid residues that do not repel, and that heterologous molecules are formed more efficiently than by using modifications which only introduce a knob and hole into CH3 as previously described.

Thus, according to findings made by the present inventors, it is possible to regulate the association of CH1 and CL. In addition, the present invention can be applied not only to regulation of the association between CH1 and CL, but also to regulation of the association between arbitrary polypeptides.

Moreover, the present inventors also confirmed that a bispecific antibody of the present invention with regulated association of the heavy chain and light chain actually retains function.

As described above, the present inventors successfully developed antigen-binding molecules in which association of the heavy chain and light chain is regulated, and thereby completed the present invention.

The present invention relates to antigen-binding molecules in which the association of the heavy chains and light chains is regulated, methods for producing an antigen-binding molecule in which the association of the heavy chains and light chains is regulated, and methods for regulating the association of the heavy chains and light chains of an antigen-binding molecule. Specifically, the present invention relates to the following:

[1] an antigen-binding molecule in which association of the heavy chain and light chain is regulated, wherein

one set or two or more sets of amino acid residues selected from the group consisting of the sets of amino acid residues shown in (a) to (c) below in the heavy chain and light chain in the antigen-binding molecule are amino acid residues that mutually repel electrically:

(a) the amino acid residue comprised in the heavy chain constant region (CH1) at position 147 as indicated by EU numbering, and the amino acid residue comprised in the light chain constant region (CL) at position 180 as indicated by EU numbering;
(b) the amino acid residue comprised in CH1 at position 147 as indicated by EU numbering, and the amino acid residue comprised in CL at position 131 as indicated by EU numbering; and,
(c) the amino acid residue comprised in CH1 at position 175 as indicated by EU numbering, and the amino acid residue comprised in CL at position 160 as indicated by EU numbering;

[2] the antigen-binding molecule of [1], further wherein amino acid residues in the set of amino acid residues shown in (d) below are amino acid residues that mutually repel electrically:

(d) the amino acid residue comprised in CH1 at position 213 as indicated by EU numbering, and the amino acid residue comprised in CL at position 123 as indicated by EU numbering;

[3] the antigen-binding molecule of [1] or [2], wherein the amino acid residues that mutually repel electrically are selected from amino acid residues comprised in either set of (X) and (Y) below:

(X) glutamic acid (E) or aspartic acid (D); and
(Y) lysine (K), arginine (R) or histidine (H);

[4] the antigen-binding molecule of any one of [1] to [3], further wherein two or more amino acid residues forming an interface between the heavy chain variable region and light chain variable region are amino acid residues that mutually repel electrically;

[5] the antigen-binding molecule of [4], wherein the amino acid residues that mutually repel electrically are one set or two sets of amino acid residues selected from the group consisting of the sets of amino acid residues shown in (a) or (b):

(a) the amino acid residue comprised in the heavy chain variable region at position 39 as indicated by Kabat numbering and the amino acid residue comprised in the light chain variable region at position 38 as indicated by Kabat numbering; or
(b) the amino acid residue comprised in the heavy chain variable region at position 45 as indicated by Kabat numbering, and the amino acid residue comprised in the light chain variable region at position 44 as indicated by Kabat numbering;

[6] the antigen-binding molecule of [4] or [5], wherein the amino acid residues that mutually repel electrically are selected from the amino acid residues comprised in either set of (X) and (Y) below:

(X) glutamic acid (E) or aspartic acid (D); and
(Y) lysine (K), arginine (R), or histidine (H);

[7] an antigen-binding molecule in which association of the heavy chain and light chain is regulated, wherein

one set or two or more sets of amino acid residues selected from the group consisting of the sets of amino acid residues shown in (a) to (c) below in the associating heavy chain and light chain in the antigen-binding molecule are amino acid residues that do not mutually repel electrically:

(a) the amino acid residue comprised in the heavy chain constant region (CH1) at position 147 as indicated by EU numbering, and the amino acid residue comprised in the light chain constant region (CL) at position 180 as indicated by EU numbering;
(b) the amino acid residue comprised in CH1 at position 147 as indicated by EU numbering, and the amino acid residue comprised in CL at position 131 as indicated by EU numbering; and,
(c) the amino acid residue comprised in CH1 at position 175 as indicated by EU numbering, and the amino acid residue comprised in CL at position 160 as indicated by EU numbering;

[8] the antigen-binding molecule of [7], further wherein amino acid residues of the set of amino acid residues shown in (d) below are amino acid residues that do not mutually repel electrically:

(d) the amino acid residue comprised in CH1 at position 213 as indicated by EU numbering, and the amino acid residue comprised in CL at position 123 as indicated by EU numbering;

[9] the antigen-binding molecule of [7] or [8], wherein the amino acid residues that do not mutually repel electrically are amino acid residues selected from each of two sets selected from the group consisting of (X) to (Z) below, and wherein the two sets are selected from among the combinations of (X) and (Y), (X) and (Z), (Y) and (Z), and (Z) and (Z):

(X) glutamic acid (E) or aspartic acid (D);
(Y) lysine (K), arginine (R) or histidine (H); and
(Z) alanine (A), asparagine (N), cysteine (C), glutamine (Q), glycine (G), isoleucine (I), leucine (L), methionine (M), phenylalanine (F), proline (P), serine (S), threonine (T), tryptophan (W), tyrosine (Y), or valine (V);

[10] the antigen-binding molecule of any one of [7] to [9], wherein the amino acid residues that do not mutually repel electrically are the amino acid residue comprised in CH1 at position 175 as indicated by EU numbering which is lysine (K); and the amino acid residues comprised in CL at position 180, position 131, and position 160 as indicated by EU numbering which are all glutamic acid (E);

[11] the antigen-binding molecule of any one of [7] to [9], wherein the amino acid residues that do not mutually repel electrically are the amino acid residues comprised in CH1 at position 147 and position 175 as indicated by EU numbering which are glutamic acid (E); and the amino acid residues comprised in CL at position 180, position 131, and position 160 as indicated by EU numbering which are all lysine (K);

[12] the antigen-binding molecule of [11], further wherein the amino acid residue comprised in CH1 at position 213 as indicated by EU numbering is glutamic acid (E), and the amino acid residue comprised in CL at position 123 as indicated by EU numbering is lysine (K);

[13] the antigen-binding molecule of any one of [7] to [12], further wherein two or more amino acid residues forming the interface between the heavy chain variable region and light chain variable region are amino acid residues that do not mutually repel electrically;

[14] the antigen-binding molecule of [13], wherein the amino acid residues that do not mutually repel electrically are one set or two sets of amino acid residues selected from the group consisting of the sets of amino acid residues shown in (a) or (b) below:

(a) the amino acid residue comprised in the heavy chain variable region at position 39 as indicated by Kabat numbering, and the amino acid residue comprised in the light chain variable region at position 38 as indicated by Kabat numbering; or
(b) the amino acid residue comprised in the heavy chain variable region at position 45 as indicated by Kabat numbering, and the amino acid residue comprised in the light chain variable region at position 44 as indicated by Kabat numbering;

[15] the antigen-binding molecule of [13] or [14], wherein the amino acid residues that do not mutually repel electrically are amino acid residues selected from each of two sets selected from the group consisting of (X) to (Z) below, and wherein the two sets are selected from among the combinations of (X) and (Y), (X) and (Z), (Y) and (Z), and (Z) and (Z):

(X) glutamic acid (E) or aspartic acid (D);
(Y) lysine (K), arginine (R) or histidine (H); and
(Z) alanine (A), asparagine (N), cysteine (C), glutamine (Q), glycine (G), isoleucine (I), leucine (L), methionine (M), phenylalanine (F), proline (P), serine (S), threonine (T), tryptophan (W), tyrosine (Y), or valine (V);

[16] the antigen-binding molecule of any one of [1] to [15], wherein the antigen-binding molecule is a bispecific antibody;

[17] a method for producing an antigen-binding molecule in which association of the heavy chain and light chain is regulated, comprising steps of (1) to (3) below:

(1) modifying nucleic acids encoding the heavy chain constant region (CH1) and the light chain constant region (CL) so that one set or two or more sets of amino acid residues selected from the group consisting of the sets of amino acid residues shown in (a) to (c) below mutually repel electrically:
(a) the amino acid residue comprised in CH1 at position 147 as indicated by EU numbering, and the amino acid residue comprised in CL at position 180 as indicated by EU numbering;
(b) the amino acid residue comprised in CH1 at position 147 as indicated by EU numbering, and the amino acid residue comprised in CL at position 131 as indicated by EU numbering; and,
(c) the amino acid residue comprised in CH1 at position 175 as indicated by EU numbering, and the amino acid residue comprised in CL at position 160 as indicated by EU numbering,
(2) introducing the modified nucleic acids into a host cell and culturing the host cell so that it expresses the nucleic acids, and
(3) collecting the antigen-binding molecule from a culture of the host cell;

[18] the method for producing an antigen-binding molecule of [17], further comprising in step (1), modifying nucleic acids so that the amino acid residues in the set of amino acid residues shown in (d) below mutually repel electrically:

(d) the amino acid residue comprised in CH1 at position 213 as indicated by EU numbering, and the amino acid residue comprised in CL at position 123 as indicated by EU numbering;

[19] the method for producing an antigen-binding molecule of [17] or [18], comprising in step (1), modifying nucleic acids so that the amino acid residues that mutually repel electrically are selected from among amino acid residues comprised in either group of (X) and (Y) below:

(X) glutamic acid (E) or aspartic acid (D); and
(Y) lysine (K), arginine (R), or histidine (H);

[20] the method for producing an antigen-binding molecule of any one of [17] to [19], further comprising in step (1), modifying nucleic acids so that two or more amino acid residues forming the interface between the heavy chain variable region and light chain variable region are amino acid residues that mutually repel electrically;

[21] the method for producing an antigen-binding molecule of [20], wherein the amino acid residues that mutually repel electrically are amino acid residues of any one set selected from the group consisting of the sets of amino acid residues shown in (a) or (b) below:

(a) the amino acid residue comprised in the heavy chain variable region at position 39 as indicated by Kabat numbering, and the amino acid residue comprised in the light chain variable region at position 38 as indicated by Kabat numbering; or
(b) the amino acid residue comprised in the heavy chain variable region at position 45 as indicated by Kabat numbering, and the amino acid residue comprised in the light chain variable region at position 44 as indicated by Kabat numbering;

[22] the method for producing an antigen-binding molecule of [20] or [21], wherein the amino acid residues that mutually repel electrically are selected from amino acid residues comprised in either set of (X) and (Y) below:

(X) glutamic acid (E) or aspartic acid (D); and
(Y) lysine (K), arginine (R), or histidine (H);

[23] a method for producing an antigen-binding molecule in which association of the heavy chain and light chain is regulated, comprising the following steps of (1) to (3):

(1) modifying nucleic acids encoding a heavy chain constant region (CH1) and a light chain constant region (CL) so that one set or two or more sets of amino acid residues selected from the group consisting of the sets of amino acid residues shown in (a) to (c) below do not mutually repel electrically:
(a) the amino acid residue comprised in the heavy chain constant region (CH1) at position 147 as indicated by EU numbering, and the amino acid residue comprised in the light chain constant region (CL) at position 180 as indicated by EU numbering;
(b) the amino acid residue comprised in CH1 at position 147 as indicated by EU numbering, and the amino acid residue comprised in CL at position 131 as indicated by EU numbering; and,
(c) the amino acid residue comprised in CH1 at position 175 as indicated by EU numbering, and the amino acid residue comprised in CL at position 160 as indicated by EU numbering,
(2) introducing the modified nucleic acids into a host cell and culturing the host cell so that it expresses the nucleic acids, and
(3) collecting the antigen-binding molecule from a culture of the host cell;

[24] the method for producing an antigen-binding molecule of [23], further comprising in step (1), modifying nucleic acids so that the amino acid residues in the set of amino acid residues shown in (d) below do not mutually repel electrically:

(d) the amino acid residue comprised in CH1 at position 213 as indicated by EU numbering, and the amino acid residue comprised in CL at position 123 as indicated by EU numbering;

[25] the method for producing an antigen-binding molecule of [23] or [24], comprising in step (1), modifying the nucleic acids so that the amino acid residues that do not mutually repel electrically are amino acids residues selected from each of two sets selected from the group consisting of (X) to (Z) below, and wherein the two sets are selected from among the combinations of (X) and (Y), (X) and (Z), (Y) and (Z), and (Z) and (Z):

(X) glutamic acid (E) or aspartic acid (D);
(Y) lysine (K), arginine (R), or histidine (H); and
(Z) alanine (A), asparagine (N), cysteine (C), glutamine (Q), glycine (G), isoleucine (I), leucine (L), methionine (M), phenylalanine (F), proline (P), serine (S), threonine (T), tryptophan (W), tyrosine (Y), or valine (V);

[26] the method for producing an antigen-binding molecule of any one of [23] to [25], comprising in step (1), modifying nucleic acids so that the amino acid residues that do not mutually repel electrically are the amino acid residue comprised in CH1 at position 175 as indicated by EU numbering which is lysine (K), and the amino acid residues comprised in CL at position 180, position 131, and position 160 as indicated by EU numbering which are all glutamic acid (E);

[27] the method for producing an antigen-binding molecule of any one of [23] to [25], comprising in step (1), modifying nucleic acids so that the amino acid residues that do not mutually repel electrically are the amino acid residues comprised in CH1 at position 147 and position 175 as indicated by EU numbering which are glutamic acid (E), and the amino acid residues comprised in CL at position 180, position 131, and position 160 as indicated by EU numbering which are all lysine (K);

[28] the method for producing an antigen-binding molecule of [27], further comprising modifying nucleic acids so that the amino acid residue comprised in CH1 at position 213 as indicated by EU numbering is glutamic acid (E), and the amino acid residue comprised in CL at position 123 as indicated by EU numbering is lysine (K);

[29] the method for producing an antigen-binding molecule of any one of [23] to [28], further comprising in step (1), modifying nucleic acids so that two or more amino acid residues forming the interface between the heavy chain variable region and light chain variable region are amino acid residues that do not mutually repel electrically;

[30] the method for producing an antigen-binding molecule of [29], wherein the amino acid residues that do not mutually repel electrically are amino acid residues of any one set selected from the group consisting of the sets of amino acid residues shown in (a) or (b) below:

(a) the amino acid residue comprised in the heavy chain variable region at position 39 as indicated by Kabat numbering, and the amino acid residue comprised in the light chain variable region at position 38 as indicated by Kabat numbering; or
(b) the amino acid residue comprised in the heavy chain variable region at position 45 as indicated by Kabat numbering, and the amino acid residue comprised in the light chain variable region at position 44 as indicated by Kabat numbering;

[31] the method for producing an antigen-binding molecule of [29] or [30], wherein the amino acid residues that do not mutually repel electrically are amino acid residues selected from each of two sets selected from the group consisting of (X) to (Z) below, and wherein the two sets are selected from among the combinations of (X) and (Y), (X) and (Z), (Y) and (Z), and (Z) and (Z):

(X) glutamic acid (E) or aspartic acid (D);
(Y) lysine (K), arginine (R) or histidine (H); and
(Z) alanine (A), asparagine (N), cysteine (C), glutamine (Q), glycine (G), isoleucine (I), leucine (L), methionine (M), phenylalanine (F), proline (P), serine (S), threonine (T), tryptophan (W), tyrosine (Y), or valine (V);

[32] an antigen-binding molecule produced according to the method for producing an antigen-binding molecule of any one of [17] to [31];

[33] the antigen-binding molecule of [32], wherein the antigen-binding molecule is a bispecific antibody;

[34] a method for regulating association of the heavy chain and light chain of an antigen-binding molecule, comprising:

modifying nucleic acids so that one set or two or more sets of amino acid residues selected from the group consisting of the sets of amino acid residues shown in (a) to (c) below are amino acid residues that mutually repel electrically:

(a) the amino acid residue comprised in CH1 at position 147 as indicated by EU numbering, and the amino acid residue comprised in CL at position 180 as indicated by EU numbering;
(b) the amino acid residue comprised in CH1 at position 147 as indicated by EU numbering, and the amino acid residue comprised in CL at position 131 as indicated by EU numbering; and
(c) the amino acid residue comprised in CH1 at position 175 as indicated by EU numbering, and the amino acid residue comprised in CL at position 160 as indicated by EU numbering;

[35] the method of [34], further comprising modifying nucleic acids so that the amino acid residues in the set of amino acid residues shown in (d) below are amino acid residues that mutually repel electrically:

(d) the amino acid residue comprised in CH1 at position 213 as indicated by EU numbering, and the amino acid residue comprised in CL at position 123 as indicated by EU numbering;

[36] the method of [34] or [35], wherein the amino acid residues that mutually repel electrically are selected from amino acid residues comprised in either set of (X) and (Y) below:

(X) glutamic acid (E) or aspartic acid (D); and
(Y) lysine (K), arginine (R), or histidine (H);

[37] the method of any one of [34] to [36], further wherein two or more amino acid residues forming the interface between the heavy chain variable region and light chain variable region are amino acid residues that mutually repel electrically;

[38] the method of [37], wherein the amino acid residues that mutually repel electrically are amino acid residues of any one set selected from the group consisting of the sets of amino acid residues shown in (a) or (b) below:

(a) the amino acid residue comprised in the heavy chain variable region at position 39 as indicated by Kabat numbering, and the amino acid residue comprised in the light chain variable region at position 38 indicated according to the Kabat numbering; or
(b) the amino acid residue comprised in the heavy chain variable region at position 45 as indicated by Kabat numbering, and the amino acid residue comprised in the light chain variable region at position 44 as indicated by Kabat numbering;

[39] the method of [37] or [38], wherein the amino acid residues that mutually repel electrically are selected from amino acid residues comprised in either set of (X) and (Y) below:

(X) glutamic acid (E) or aspartic acid (D); and
(Y) lysine (K), arginine (R), or histidine (H);

[40] a method for regulating association of the heavy chain and light chain of an antigen-binding molecule, comprising:

modifying nucleic acids so that one set or two or more sets of amino acid residues selected from the group consisting of the sets of amino acid residues shown in (a) to (c) below are amino acid residues that do not mutually repel electrically:

(a) the amino acid residue comprised in CH1 at position 147 as indicated by EU numbering, and the amino acid residue comprised in CL at position 180 as indicated by EU numbering;
(b) the amino acid residue comprised in CH1 at position 147 as indicated by EU numbering, and the amino acid residue comprised in CL at position 131 as indicated by EU numbering; and,
(c) the amino acid residue comprised in CH1 at position 175 as indicated by EU numbering, and the amino acid residue comprised in CL at position 160 as indicated by EU numbering;

[41] the method of [40], further comprising modifying nucleic acids so that the amino acid residues in the set of amino acid residues shown in (d) below are amino acid residues that do not mutually repel electrically:

(d) the amino acid residue comprised in CH1 at position 213 as indicated by EU numbering, and the amino acid residue comprised in CL at position 123 as indicated by EU numbering;

[42] the method of [40] or [41], wherein the amino acid residues that do not mutually repel electrically are amino acid residues selected from each of two sets selected from the group consisting of (X) to (Z) below, and wherein the two sets are selected from among the combinations of (X) and (Y), (X) and (Z), (Y) and (Z), and (Z) and (Z):

(X) glutamic acid (E) or aspartic acid (D);
(Y) lysine (K), arginine (R), or histidine (H); and
(Z) alanine (A), asparagine (N), cysteine (C), glutamine (Q), glycine (G), isoleucine (I), leucine (L), methionine (M), phenylalanine (F), proline (P), serine (S), threonine (T), tryptophan (W), tyrosine (Y), or valine (V);

[43] the method of any one of [40] to [42], wherein the amino acid residues that do not mutually repel electrically are the amino acid residue comprised in CH1 at position 175 as indicated by EU numbering which is lysine (K), and the amino acid residues comprised in CL at position 180, position 131, and position 160 as indicated by EU numbering which are all glutamic acid (E);

[44] the method of any one of [40] to [42], wherein the amino acid residues that do not mutually repel electrically are the amino acid residues comprised in CH1 at position 147 and position 175 as indicated by EU numbering which are glutamic acid (E), and the amino acid residues comprised in CL at position 180, position 131, and position 160 as indicated by EU numbering which are all lysine (K);

[45] the method of [44], further wherein the amino acid residue contained in CH1 at position 213 as indicated by EU numbering is glutamic acid (E), and the amino acid residue contained in CL at position 123 as indicated by EU numbering is lysine (K);

[46] the method of any one of [34] to [45], wherein the antigen-binding molecule is a bispecific antibody;

[47] a composition containing the antigen-binding molecule of any one of [1] to [16], [32], and [33], and a pharmaceutically acceptable carrier;

[48] a nucleic acid encoding the antigen-binding molecule of any one of [1] to [16], [32], and [33]; and

[49] a host cell having the nucleic acid of [48].

BRIEF DESCRIPTION OF THE DRAWINGS

FIG. 1 is a model diagram of an CH1/CL interface.

FIG. 2 is a conceptual antibody showing possible combinations of H chain and L chain when the antibody is prepared by mixing one type of H chain and two types of L chains. Mutated sites which gave a large proportion of the antibody with the combination of E and K as shown in the box are thought to interact electrically.

FIG. 3 depicts graphs showing results of the AIEX analysis of each of the antibodies.

FIG. 4 depicts graphs showing results of the AIEX analysis of each of the antibodies.

FIG. 5 depicts graphs showing results of the AIEX analysis of each of the antibodies.

FIG. 6 depicts a graph showing results of the AIEX analysis of each of the antibodies.

FIG. 7 depicts graphs showing results of the CIEX analysis of each of the antibodies.

FIG. 8 depicts graphs showing results of the CIEX analysis of each of the antibodies.

FIG. 9-1 depicts graphs showing the results of CIEX analysis of each of the antibodies. FIG. 9-2 is a continuation of FIG. 9-1.

FIG. 10 depicts graphs showing results of the CIEX analysis of each of the antibodies.

FIG. 11 depicts graphs showing results of the CIEX analysis of each of the antibodies.

FIG. 12 is a diagram comparing the H chain CH1 by aligning the amino acid sequences of human IgA1 (SEQ ID NO: 63), IgA2 (SEQ ID NO: 64), IgD (SEQ ID NO: 65), IgE (SEQ ID NO: 66), IgG1 (SEQ ID NO: 67), IgG2 (SEQ ID NO: 68), IgG3 (SEQ ID NO: 69), IgG4 (SEQ ID NO: 70), and IgM (SEQ ID NO: 71); and the L chain CL by aligning the amino acid sequences of human IgK (Kappa) (SEQ ID NO: 72), IgL1 (SEQ ID NO: 73), IgL2 (SEQ ID NO: 74), IgL3 (SEQ ID NO: 75), IgL6 (SEQ ID NO: 76), IgL7 (SEQ ID NO: 77) (Lambda).

MODE FOR CARRYING OUT THE INVENTION

The present invention relates to antibodies in which the association of the heavy chains and light chains is regulated, methods for producing an antibody in which the association of the heavy chains and light chains is regulated, and methods for regulating the association of the heavy chains and light chains of an antibody.

In the present invention, the term “antibody” is used synonymously with “antigen-binding molecule”. That is, in the present invention, the terms “antibody” and “antigen-binding molecule” are used in the broadest sense, and include monoclonal antibodies, polyclonal antibodies, and antibody variants (such as chimeric antibodies, humanized antibodies, low molecular weight antibodies (including antibody fragments to which other molecules may be added arbitrarily), and polyspecific antibodies) provided that they demonstrate the desired antigen-binding activity or biological activity. An example of an “antibody” or “antigen-binding molecule” in the present invention is a molecule in which an HAS-binding scaffold has been added to the Fab (an antibody in which only the Fab portion is normal). In addition, in the present invention, an “antibody” may also be a polypeptide or a heteromeric multimer. Preferred antibodies are monoclonal antibodies, chimeric antibodies, humanized antibodies, human antibodies, Fc-fusion antibodies and low molecular weight antibodies such as antibody fragments.

The antibody of the present invention is an antibody in which the association of the heavy chain and light chain is regulated, in which the heavy chain and light chain constituting the antibody are a combination of heavy chain and light chain of interest, and in which the amino acid residues at given locations in the constant region of the heavy chain (CH1) and the constant region of the light chain are mutually electrically repelling amino acid residues (having the same charge).

In the present invention, by making amino acid residues at given locations in the constant region of the heavy chain (CH1) and constant region of the light chain of an undesired combination of heavy chain and light chain into amino acid residues that mutually repel electrically (i.e., that have the same charge), the formation of undesired combinations of heavy chain and light chain can be prevented by utilizing this charge repulsion, and as a result, the desired combination of heavy chain and light chain can be formed.

In another embodiment, the antibody of the present invention is an antibody in which the association of the heavy chain and light chain is regulated, in which the heavy chain and light chain constituting the antibody associate as a combination of heavy chain and light chain of interest, and in which the amino acid residues at given locations in the constant region of the heavy chain (CH1) and in the constant region of the light chain do not mutually repel electrically.

By making amino acid residues at given locations in the heavy chain constant region (CH1) and the light chain constant region of a desired combination of heavy chain and light chain into amino acid residues that do not mutually repel electrically, a desired combination of heavy chain and light chain can be formed, for example, by using the attractive force of the electric charges. In the present invention, the term “polypeptide” generally refers to peptides and proteins whose length is about ten amino acids or longer. Polypeptides are ordinarily derived from organisms but are not particularly limited thereto, and for example, they may be composed of an artificially designed sequence. They may also be any of naturally derived polypeptides, synthetic polypeptides, recombinant polypeptides, or such. Additionally, fragments of the above-mentioned polypeptides are also included in the polypeptides of the present invention.

In the present invention, the phrases “to regulate association” and “association is regulated” refer to regulating to achieve a desired association condition, and more specifically refers to regulating so that undesirable associations are not formed between the heavy chain and light chain.

In the present invention, the term “interface” generally refers to the association surface that results from association (interaction), and amino acid residues that form the interface are ordinarily one or more amino acid residues included in the polypeptide regions which participate in the association, and are more preferably amino acid residues that approach each other during association and are involved in the interaction. More specifically, this interaction includes, for example, instances where the amino acid residues come close during the association to form hydrogen bonds, electrostatic interactions, or salt bridges with each other.

In the present invention, the phrase, “amino acid residues forming an interface” more specifically refers to amino acid residues included in the polypeptide region that constitutes the interface. For example, polypeptide regions constituting the interface refer to polypeptide regions responsible for selective binding between molecules such as in antibodies, ligands, receptors, or substrates. More specifically, in antibodies, such examples include heavy chain constant regions, heavy chain variable regions, light chain constant regions, and light chain variable regions.

“Modification” of amino acid residues in the present invention specifically refers to substituting original amino acid residue(s) for other amino acid residue(s), deleting original amino acid residue(s), adding new amino acid residue(s), and such, but preferably refers to substituting one or more original amino acid residues for other amino acid residues.

In a preferred embodiment of the antibody of the present invention, the antibody has amino acid residues at given locations in the heavy chain constant region (CH1) and light chain constant region of an undesired combination of heavy chain and light chain before association regulation which electrically repel (which have the same charge).

By modifying amino acid residues in the aforementioned antibody into amino acid residues that mutually repel electrically (have the same charge), association of these amino acid residues is thought to be inhibited by the repulsive force of electrical charges.

In another preferred embodiment of the antibody of the present invention, the antibody has amino acid residues involved in association at the interface of polypeptides that do not mutually repel electrically.

In the aforementioned antibody, by modifying amino acid residues involved in association at the interface of polypeptides into amino acid residues that do not mutually repel electrically, association of these amino acid residues is thought to be promoted by, for example, the attractive force of their electrical charges.

Thus, in the aforementioned antibody, the modified amino acid residues are preferably amino acid residues that approach each other at association, in the polypeptide regions forming the interface.

The amino acid residues that approach during association can be determined by, for example, analyzing the three-dimensional structure of a polypeptide, and investigating the amino acid sequences of the polypeptide regions that form an interface during polypeptide association. Amino acid residues at the interface that mutually approach each other are preferable targets of “modification” in the antibody of the present invention.

Some amino acids are known to be electrically charged. In general, lysine (K), arginine (R) and histidine (H) are known to be amino acids having a positive charge (positively charged amino acids). Aspartic acid (D), glutamic acid (E), and such are known to be amino acids having a negative charge (negatively charged amino acids). In addition, alanine (A), asparagine (N), cysteine (C), glutamine (Q), glycine (G), isoleucine (I), leucine (L), methionine (M), phenylalanine (F), proline (P), serine (S), threonine (T), tryptophan (W), tyrosine (Y), valine (V), and the like are known to be amino acids that do not have a charge, or nonpolar amino acids.

Thus, amino acids that mutually repel electrically (have the same charge) in the present invention refer to:

(1) amino acids in which one of the amino acids is a positively charged amine acid and the other amino acid is also a positively charged amino acid, and
(2) amino acids in which one of the amino acids is a negatively charged amino acid and the other amino acid is also a negatively charged amino acid.

Further, amino acids that do not mutually repel electrically in the present invention refer to:

(1) amino acids in which one of the amino acids is a positively charged amino acid and the other amino acid is a negatively charged amino acid,
(2) amino acids in which one of the amino acids is a positively charged amino acid and the other amino acid is an uncharged amino acid or a nonpolar amino acid,
(3) amino acids in which one of the amino acids is a negatively charged amino acid and the other amino acid is an uncharged amino acid or a nonpolar amino acid, and
(4) amino acids in which both of the amino acids are uncharged amino acids or nonpolar amino acids.

Amino acids can be modified according to various methods known in the field of the art. Examples of these methods include, but are not limited to site-directed mutagenesis (Hashimoto-Gotoh, T., Mizuno, T., Ogasahara, Y. and Nakagawa, M. (1995) An oligodeoxyribonucleotide-directed dual amber method for site-directed mutagenesis, Gene 152, 271-275; Zoller, M. J. and Smith, M. (1983) Oligonucleotide-directed mutagenesis of DNA fragments cloned into M13 vectors, Methods Enzymol. 100, 468-500; Kramer, W., Drutsa, V., Jansen, H. W., Kramer, B., Pflugfelder, M. and Fritz, H. J. (1984) The gapped duplex DNA approach to oligonucleotide-directed mutation construction, Nucleic Acids Res. 12, 9441-9456; Kramer, W. and Fritz, H. J. (1987) Oligonucleotide-directed construction of mutations via gapped duplex DNA, Methods Enzymol. 154, 350-367; Kunkel, T. A. (1985) Rapid and efficient site-specific mutagenesis without phenotypic selection, Proc. Natl. Acad. Sci. USA 82, 488-492), PCR mutagenesis, cassette mutagenesis, etc.

Examples of amino acid modifications include modification of an uncharged amino acid or a nonpolar amino acid into a positively charged amino acid, modification of an uncharged amino acid or a nonpolar amino acid into a negatively charged amino acid, modification of a positively charged amino acid into a negatively charged amino acid, and modification of a negatively charged amino acid into a positively charged amino acid. Furthermore, modification of an uncharged amino acid or a nonpolar amino acid into a different uncharged or nonpolar amino acid, modification of a positively charged amino acid into a different positively charged amino acid, and modification of a negatively charged amino acid into a different negatively charged amino acid are also included in the amino acid modifications of the present invention.

Modifying amino acids in the present invention includes making one modification in each of the heavy and light chain, or making multiple modifications to each of the heavy and light chain. In addition, the number of modifications added to the heavy chain and light chain may be the same or different.

Modifying amino acids sin the present invention includes making multiple modifications into positively charged amino acids on either the heavy chain or light chain, and making multiple modifications into negatively charged amino acids on the other chain. Moreover, multiple modifications into positively charged amino acids as well as multiple modifications into negatively charged amino acids may be made on the same heavy chain or light chain. In these modifications, modifications into uncharged amino acids or nonpolar amino acids as well as modifications of uncharged amino acids or nonpolar amino acids may also be suitably combined.

In the modifications of the present invention, for example, the amino acids on one of the chains can be used as they are without being modified s, and in such cases, the heavy chain and light chain do not need to be both modified, and only one of the chains may be modified.

Although there are no particular limitations to the number of amino acid residues subjected to modification in the antibody of the present invention, for example, when modifying the constant region of the antibody, in order not to reduce the binding activity toward the antigen and not to increase immunogenicity, it is preferable to modify as few amino acid residues as possible. The aforementioned “few” refers to, for example, a number of about 1 to 30, preferably a number of about 1 to 20, even more preferably a number of about 1 to 15, and most preferably a number of 1 to 5.

In the present invention, the term “antibody” is used in the broadest sense, and includes monoclonal antibodies, polyclonal antibodies, antibody variants (such as chimeric antibodies, humanized antibodies, low molecular weight antibodies (including antibody fragments), and polyspecific antibodies) as long as they demonstrate the desired biological activity. In addition, the “antibody” in the present invention may be either a polypeptide or a heteromeric multimer. Preferred antibodies are monoclonal antibodies, chimeric antibodies, humanized antibodies, human antibodies, Fc-fusion antibodies, and low molecular weight antibodies such as antibody fragments.

In the context of the present invention, the term “multispecific antibody” (used in the present description to have the same meaning as “polyspecific antibody”) refers to an antibody that may bind specifically to different types of epitopes. More specifically, multispecific antibodies are antibodies having specificity to at least two different types of epitopes, and, in addition to antibodies recognizing different antigens, antibodies recognizing different epitopes on the same antigen are also included. (For example, when the antigens are heterologous receptors, multispecific antibodies can recognize different domains constituting the heterologous receptors; alternatively, when the antigens are monomers, multispecific antibodies recognize multiple sites on the monomer antigens.) Ordinarily, such molecules bind to two antigens (bispecific antibodies; used in the present description to have the same meaning as “dual-specific antibodies”), but they may even have specificity toward more antigens (for example three types).

In addition to the antibodies described above, the antibodies of the present invention include antibodies whose amino acid sequences have been modified by amino acid substitutions, deletions, additions, and/or insertions, or chimerization, humanization, and such. Such amino acid sequence modifications, such as amino acid substitutions, deletions, additions, and/or insertions, and humanization and chimerization, can be achieved by methods known to those skilled in the art. When the antibodies of the present invention are prepared as recombinant antibodies, likewise, the amino acid sequences of the antibody variable and constant regions may also be modified by amino acid substitutions, deletions, additions, and/or insertions, or chimerization, humanization and the like.

The antibodies of the present invention may be derived from any animal, such as a mouse, human, rat, rabbit, goat, or camel. Furthermore, the antibodies may be modified, for example, chimeric antibodies, and in particular, modified antibodies that include amino acid substitutions in their sequence, such as humanized antibodies. The antibodies may be any type of antibody, such as antibody modification products linked with various molecules, antibody fragments, and low molecular weight antibodies.

“Chimeric antibodies” are antibodies prepared by combining sequences derived from different animals. An example is an antibody having heavy and light chain variable (V) regions from a mouse antibody and heavy and light chain constant (C) regions from a human antibody. Chimeric antibodies can be prepared by known methods. To obtain such chimeric antibodies, for example, a DNA encoding an antibody V region may be ligated with a DNA encoding a human antibody constant region; the resulting ligation product can be inserted into an expression vector; and the construct can be introduced into a host to produce the chimeric antibody.

“Humanized antibodies” are also referred to as reshaped human antibodies, and can be obtained by substituting the complementarity determining region (CDR) of a human antibody for the CDR of an antibody derived from a nonhuman mammal, for example, a mouse. Methods for identifying CDRs are known in the art (Kabat et al., Sequence of Proteins of Immunological Interest (1987), National Institute of Health, Bethesda, Md.; Chothia et al., Nature (1989) 342:877). General genetic recombination techniques suitable for this purpose are also known (see European Patent Application Publication No. EP 125023; and WO 96/02576). For example, the CDR of a mouse antibody can be determined by known methods, and a DNA can be prepared such that it encodes an antibody in which the CDR is ligated with the framework region (FR) of a human antibody. A humanized antibody can then be produced using a system that uses conventional expression vectors. Such DNAs can be synthesized by PCR, using as primers several oligonucleotides designed to include portions that overlap the ends of both the CDR and FR regions (see the method described in WO 98/13388). Human antibody FRs linked via CDRs are selected such that the CDRs form a suitable antigen binding site. If required, amino acids in the FRs of an antibody variable region may be modified so that the CDRs of the reshaped human antibody can form a suitable antigen binding site (Sato, K. et al., Cancer Res. (1993) 53:851-856). Modifiable amino acid residues in the FRs include portions that directly bind to an antigen via non-covalent bonds (Amit et al., Science (1986) 233: 747-53), portions that have some impact or effect on the CDR structure (Chothia et al., J. Mol. Biol. (1987) 196: 901-17), and portions involved in the interaction between VH and VL (EP 239400).

The heavy chain constant region of the antibody of the present invention is preferably a human heavy chain constant region. In addition, examples of antibody heavy chain constant regions include IgA1, IgA2, IgD, IgE, IgG1, IgG2, IgG3, IgG4 and IgM type constant regions. The heavy-chain constant region of the antibody of the present invention is preferably an IgG1 type constant region, and particularly preferably a human IgG1 constant region, but it is not limited thereto. Several allotype sequences obtained by genetic polymorphism are described in Sequences of Proteins of Immunological Interest, NIH Publication No. 91-3242 as human IgG1 constant region, and any of these may be used in the present invention.

Moreover, the light chain constant region of the antibody of the present invention is preferably a human light chain constant region. Examples of antibody light chain constant region include IgK (Kappa), IgL1, IgL2, IgL3, IgL6 and IgL7 (Lambda) type constant regions. The light chain constant region of the antibody of the present invention is preferably a human IgK (Kappa) constant region, but is not limited thereto. The amino acid sequence of the human IgK (Kappa) constant region is known (SEQ ID NO: 72). Several allotype sequences obtained by genetic polymorphism are described in Sequences of Proteins of Immunological Interest, NIH Publication No. 91-3242 as human IgK (Kappa) constant region and human IgL7 (Lambda) constant region, and any of these may be used in the present invention.

Antibody constant regions, in particular, heavy chain constant regions, may be modified as necessary in order to improve antibody function or antibody stability. Examples of modifications for improving antibody function include modifications that strengthen or weaken the binding between an antibody and an Fcγ receptor (FcγR), modifications that strengthen or weaken the binding between an antibody and FcRn, modifications that strengthen or weaken antibody cytotoxic activity (such as ADCC activity and CDC activity), and such. In addition, modifications that improve antibody heterogeneity and modifications that improve immunogenicity and/or pharmacokinetics may also be included.

Moreover, as the heterogeneity of the heavy chain C-terminal sequence of the IgG antibody, amidation of the C-terminal carboxyl group by deletion of the C-terminal amino acid, lysine residue, or by deletion of the two C-terminal amino acids, glycine and lysine, has been reported the (Anal. Biochem. 2007 Jan. 1:360(1):75-83). Thus, in the present invention, to lower heterogeneity of the heavy chain C terminus, it is preferable to use an IgG in which the C-terminal lysine or the C-terminal lysine and glycine have been deleted.

Since their antigenicity in the human body has been attenuated, chimeric and humanized antibodies using human-derived sequences are expected to be useful when administered to humans for therapeutic purposes or such.

Moreover, low molecular weight antibodies are useful as the antibodies because of their in vivo kinetic characteristics and low-cost production using E. coli, plant cells, or such.

Antibody fragments are one type of low molecular weight antibody. The term “low molecular weight antibody” includes antibodies that include an antibody fragment as a partial structural unit. The low molecular weight antibodies of the present invention are not particularly limited by their structure nor their method of production, so long as they have antigen binding activity. Some low molecular weight antibodies have an activity greater than that of a whole antibody (Orita et al., Blood (2005) 105:562-566). Herein, the “antibody fragments” are not particularly limited, so long as they are a portion of a whole antibody (for example, whole IgG). However, the antibody fragments preferably include a heavy chain variable region (VH) or a light chain variable region (VL), and further include CH1 or CL. Examples of preferred antibody fragments are: Fab, F(ab′)2, and Fab′. The amino acid sequences of a VH, VL, CH1, and CL in an antibody fragment may be modified by substitution, deletion, addition, and/or insertion. Furthermore, some portions of a CH1, CL, VH, and VL may be deleted, so long as the resulting fragments retain their antigen binding ability, and antibody fragments such as scFv, Fab, domain antibody (dAb), and VHH, HAS binding scaffold, PEG, albumin, cytokines, toxins, and the like (the molecules described in Biodrugs, 2009, 23(2):93-109; Methods Mol. Med., 2005, 109:347-74; AAPS J., 2006 Aug. 18, 8(3):E532-51; etc.) may also be added to increase the pharmacokinetics (PK) or drug efficacy.

An antibody fragment can be prepared by treating an antibody with an enzyme, for example, a protease such as papain or pepsin (see Morimoto et al., J. Biochem. Biophys. Methods (1992) 24: 107-17; Brennan et al., Science (1985) 229:81). Alternatively, antibody fragments can also be produced by genetic recombination based on their amino acid sequence.

A low molecular weight antibody having a structure that results from modification of an antibody fragment can be prepared using antibody fragments obtained by enzyme treatment or genetic recombination. Alternatively, after constructing a gene which encodes a whole low molecular weight antibody, and introducing the construct into an expression vector, the low molecular weight antibody may be expressed in appropriate host cells (see, for example, Co et al., J. Immunol. (1994) 152: 2968-76; Better and Horwitz, Methods Enzymol. (1989) 178: 476-96; Pluckthun and Skerra, Methods Enzymol. (1989) 178: 497-515; Lamoyi, Methods Enzymol. (1986) 121: 652-63; Rousseaux et al., Methods Enzymol. (1986) 121: 663-9; Bird and Walker, Trends Biotechnol. (1991) 9: 132-7).

A preferred example of the antibody of the present invention is a heteromeric multimer having two or more types of CH1 and two or more types of CL. This heteromeric multimer preferably recognizes two or more types of epitopes, and an example thereof is a polyspecific antibody.

A preferred example of a polyspecific antibody of the present invention is a bispecific antibody. Thus, an example of a preferred embodiment of the antibody of the present invention is a bispecific antibody composed of two types of heavy chains (a first heavy chain and a second heavy chain) and two types of light chains (a first light chain and a second light chain).

Describing the “bispecific antibodies” of the preferred embodiments of the antibodies of the present invention more precisely, the above-mentioned “first heavy chain” refers to one of the two heavy chains (H chains) forming the antibody, and the “second H chain” refers to the other H chain that is different from the first H chain. That is, of the two H chains, one of them can be arbitrarily defined as the first H chain and the other can be defined as the second H chain. Similarly, the “first light chain” refers to one of the two light chains (L chains) forming the bispecific antibody, and the “second L chain” refers to the other L chain that is different from the first L chain. Of the two L chains, one of them can be arbitrarily defined as the first L chain and the other can be defined as the second L chain. Ordinarily, the first L chain and the first H chain are derived from a same antibody that recognizes a certain antigen (or epitope), and the second L chain and the second H chain are also derived from a same antibody that recognizes a certain antigen (or epitope). Herein, the L chain-H chain pair formed by the first H chain and L chain is called the first pair, and the L chain-H chain pair formed by the second H chain and L chain is called the second pair. The antigen (or epitope) used to produce the antibody from which the second pair derives is preferably different from the antigen used to produce the antibody from which the first pair derives. More specifically, antigens recognized by the first pair and the second pair may be the same, but preferably, the pairs recognize different antigens (or epitopes). In this case, the H chains and L chains of the first pair and second pair preferably have amino acid sequences that differ from each other. When the first pair and the second pair recognize different epitopes, the first pair and the second pair may recognize a completely different antigen, or they may recognize different sites (different epitopes) on the same antigen. Furthermore, one of them may recognize an antigen such as a protein, peptide, gene, or sugar, and the other may recognize cytotoxic substances such as radioactive substances, chemotherapeutic agents, or cell-derived toxins. However, when one wishes to produce an antibody having pairs formed by specific combinations of H chains and L chains, those specific H chains and L chains may be arbitrary determined to be the first pair and second pair.

As for the genes encoding the H chain or L chain of antibodies before introduction of mutations in the present invention (herein, it may be simply referred to as “an antibody of the present invention”), known sequences can be used, or they can be obtained by methods known to those skilled in the art. For example, they may be obtained from an antibody library, or they may be obtained by cloning genes encoding the antibody from hybridomas producing monoclonal antibodies.

Regarding antibody libraries, many antibody libraries are already well known, and since methods for producing antibody libraries are known, those skilled in the art can appropriately obtain antibody libraries. For example, regarding antibody phage libraries, one can refer to the literature such as Clackson et al., Nature 1991, 352: 624-8; Marks et al., J. Mol. Biol. 1991, 222: 581-97; Waterhouses et al., Nucleic Acids Res. 1993, 21: 2265-6; Griffiths et al., EMBO J. 1994, 13: 3245-60; Vaughan et al., Nature Biotechnology 1996, 14: 309-14; and Japanese Patent Kohyo Publication No. (JP-A) H10-504970 (unexamined Japanese national phase publication corresponding to a non-Japanese international publication). In addition, known methods, such as methods that use eukaryotic cells as libraries (WO95/15393) and ribosome display methods, may be used. Furthermore, techniques to obtain human antibodies by panning using human antibody libraries are also known. For example, variable regions of human antibodies can be expressed on the surface of phages as single chain antibodies (scFvs) using phage display methods, and phages that bind to antigens can be selected. Genetic analysis of the selected phages can determine the DNA sequences encoding the variable regions of human antibodies that bind to the antigens. Once the DNA sequences of scFvs that bind to the antigens is revealed, suitable expression vectors can be produced based on these sequences to obtain human antibodies. These methods are already well known, and one can refer to WO92/01047, WO92/20791, WO93/06213, WO93/11236, WO93/19172, WO95/01438, and WO95/15388.

As for methods for obtaining genes encoding antibodies from hybridomas, known techniques may be basically used, which involve using of desired antigens or cells expressing the desired antigens as sensitizing antigens, using these to perform immunizations according to conventional immunization methods, fusing the immune cells thus obtained with known parent cells by ordinary cell fusion methods, screening monoclonal antibody producing cells (hybridomas) by ordinary screening methods, synthesizing cDNAs of antibody variable regions (V regions) from mRNAs of the obtained hybridomas using reverse transcriptase, and linking them with DNAs encoding the desired antibody constant regions.

The sensitizing antigens for obtaining the aforementioned antibody genes encoding the H chain and L chain are not particularly limited to the examples described below, but include both complete antigens having immunogenicity and incomplete antigens including haptens and the like that do not demonstrate immunogenicity. There are no particular limitations on the antigen for the antibodies of the present invention, and for example, a full-length protein or a partial peptide of a target protein, as well as substances composed of polysaccharides, nucleic acids, lipids, and the like that are known to be able to serve as an antigen can be used. Antigens can be prepared in accordance with methods that are known to those skilled in the art, such as methods using baculoviruses (such as that described in WO 98/46777). Hybridomas can be produced, for example, following methods of Milstein et al. (G. Kohler and C. Milstein, Methods Enzymol. 1981, 73: 3-46), and such. When the immunogenicity of an antigen is low, it can be linked to a macromolecule that has immunogenicity, such as albumin, and then used for immunization. Furthermore, by linking antigens with other molecules as necessary, they can be converted into soluble antigens. When transmembrane molecules such as receptors are used as antigens, portions of the extracellular regions of the receptors can be used as a fragment, or cells expressing transmembrane molecules on their cell surface may be used as immunogens.

Antibody-producing cells can be obtained by immunizing animals using suitable sensitizing antigens described above. Alternatively, antibody-producing cells can be prepared by in vitro immunization of lymphocytes that can produce antibodies. Various mammals can be used as the animals for immunization, and rodents, lagomorphas and primates are generally used. Specific examples of such animals include mice, rats, and hamsters for rodents, rabbits for lagomorphas, and monkeys including the cynomolgus monkey, rhesus monkey, hamadryas, and chimpanzees for primates.

Transgenic animals carrying human antibody gene repertoires are also known, and human antibodies can be obtained by using these animals (see WO96/34096; Mendez et al., Nat. Genet. 1997, 15: 146-56). Instead of using such transgenic animals, for example, desired human antibodies having binding activity against antigens can be obtained by in vitro sensitization of human lymphocytes with desired antigens or cells expressing the desired antigens, and then fusing the sensitized lymphocytes with human myeloma cells such as U266 (see Japanese Patent Application Kokoku Publication No. (JP-B) H1-59878 (examined, approved Japanese patent application published for opposition)). Furthermore, desired human antibodies can be obtained by immunizing transgenic animals carrying a complete repertoire of human antibody genes with desired antigens (see WO93/12227, WO92/03918, WO94/02602, WO96/34096, and WO96/33735).

Animal immunization is carried out by appropriately diluting and suspending a sensitizing antigen in Phosphate-Buffered Saline (PBS), physiological saline, or such, forming an emulsion by mixing an adjuvant if necessary, and intraperitoneally or subcutaneously injecting this into animals. After that, the sensitizing antigen mixed with Freund's incomplete adjuvant is preferably administered several times every four to 21 days. Antibody production can be confirmed by measuring the target antibody titer in animal sera using conventional methods.

Antibody-producing cells obtained from lymphocytes or animals immunized with a desired antigen can be fused with myeloma cells to generate hybridomas using conventional fusing agents (for example, polyethylene glycol) (Goding, Monoclonal Antibodies: Principles and Practice, Academic Press, 1986, 59-103). Then, as required, hybridoma cells can be cultured and grown, and the binding specificity, affinity, or activity of the antibody produced from these hybridomas can be measured using known analysis methods, such as immunoprecipitation, radioimmunoassay (RIA), and enzyme-linked immunosorbent assay (ELISA). Thereafter, hybridomas that produce antibodies of interest whose binding specificity, affinity, or activity has been determined can be subcloned by methods such as limiting dilution.

Next, genes encoding the antibodies of interest can be cloned from hybridomas or antibody-producing cells (sensitized lymphocytes, and such) using probes that may specifically bind to the antibodies (for example, oligonucleotides complementary to sequences encoding the antibody constant regions). Cloning from mRNAs using RT-PCR is also possible. Immunoglobulins are classified into five different classes, IgA, IgD, IgE, IgG, and IgM. These classes are further divided into several subclasses (isotypes) (for example, IgG-1, IgG-2, IgG-3, and IgG-4; IgA-1 and IgA-2; and such). H chains and L chains used in the present invention to produce antibodies are not particularly limited and may derive from antibodies belonging to any of these classes or subclasses; however, IgG is particularly preferred.

Herein, it is possible to modify H-chain-encoding genes and L-chain-encoding genes using genetic engineering techniques. Genetically modified antibodies, such as chimeric antibodies and humanized antibodies, that have been artificially modified for the purpose of decreasing heterologous antigenicity and such against humans, can be appropriately produced for antibodies such as mouse antibodies, rat antibodies, rabbit antibodies, hamster antibodies, sheep antibodies, and camel antibodies.

Chimeric antibodies are antibodies composed of a nonhuman mammal antibody H chain and L chain variable regions, such as those of a mouse antibody, and the H chain and L chain constant regions of a human antibody. They can be obtained by ligating the DNA encoding a variable region of a mouse antibody to the DNA encoding a constant region of a human antibody, incorporating them into an expression vector, and introducing the vector into a host for production of the antibody. A humanized antibody is also called a reshaped human antibody. This humanized antibody can be synthesized by PCR from a number of oligonucleotides produced so that they have overlapping portions at the ends of DNA sequences designed to link the complementarity determining regions (CDRs) of an antibody of a nonhuman mammal (such as a mouse). The obtained DNA can be ligated to a DNA encoding a human antibody constant region. The ligated DNA can be incorporated into an expression vector, and the vector can be introduced into a host to produce the antibody (see EP239400 and WO96/02576). Human antibody FRs that are ligated via the CDRs are selected when the CDRs form a favorable antigen-binding site. If necessary, amino acids in the framework region of an antibody variable region may be substituted such that the CDRs of the reshaped human antibody form an appropriate antigen-binding site (K. Sato et al., Cancer Res. 1993, 53: 851-856).

In addition to the humanization techniques described above, antibodies may be modified to improve their biological properties, for example, antigenic affinity. Such modifications can be carried out using methods such as site-directed mutagenesis (see for example, Kunkel (1985) Proc. Natl. Acad. Sci. USA 82: 488), PCR mutagenesis, and cassette mutagenesis. In general, mutant antibodies whose biological properties have been improved show amino acid sequence homology and/or similarity of 70% or higher, more preferably 80% or higher, and even more preferably 90% or higher (for example, 95% or higher, 97%, 98%, 99%, etc.), when compared to the amino acid sequence of the original antibody variable region. Herein, a sequence homology and/or similarity is defined as the ratio of amino acid residues that are homologous (same residue) or similar (amino acid residues classified into the same group based on the general properties of amino acid side chains) to the original antibody residues, after the sequence homology value has been maximized by sequence alignment and gap introduction as necessary. Generally, naturally-occurring amino acid residues are classified into groups based on the characteristics of their side chains:

(1) hydrophobic: alanine, isoleucine, norleucine, valine, methionine, and leucine;
(2) neutral hydrophilic: asparagine, glutamine, cysteine, threonine, and serine;
(3) acidic: aspartic acid, and glutamic acid;
(4) basic: arginine, histidine, and lysine;
(5) residues that affect the orientation of the chain: glycine, and proline; and
(6) aromatic: tyrosine, tryptophan, and phenylalanine.

Normally, an antigen binding site of an antibody is formed by the interactions of a total of six complementarity determining regions (hypervariable portions; CDRs) present in the variable regions of the H chain and L chain. It is known that even one of these variable regions has the ability to recognize and bind the antigen, although the affinity will be lower than that of the variable regions containing all the binding sites. Thus, with regard to the antibody genes of the present invention encoding H chains and L chains, the polypeptides encoded by these genes are only required to maintain the ability of binding to a desired antigen and to encode a fragment portion containing the respective antigen-binding sites of the H chain and L chain.

A more detailed explanation is provided below on the case of an IgG-type bispecific antibody having two types of heavy chain constant regions CH1 (CH1-A and CH1-B) and two types of light chain constant regions (CL-A and CL-B); however, the present invention can be similarly applied to other antibodies as well.

When one wishes to obtain a bispecific antibody that would recognize one epitope by the first CH1-A and the first CL-A, and another epitope by the second CH1-B and the second CL-B, theoretically there is the possibility that 10 types of antibody molecules may be produced when each of the four types of chains is expressed for producing that antibody.

In this case, desired antibody molecules can be preferentially acquired if, for example, the association is regulated so that association of CH1-A and CL-B and/or between CH1-B and CL-A is inhibited.

An example is modifying amino acid residues forming an interface between CH1-A and CL-B into positively charged amino acid residues and modifying amino acid residues forming an interface between CH1-B and CL-A into negatively charged amino acid residues. As a result of these modifications, unintended association between CH1-A and CL-B is inhibited since the amino acid residues forming the interface are both positively charged, and association between CH1-B and CL-A is also inhibited since the amino acid residues forming the interface are both negatively charged. Thus, the unintended association between CH1-A and CL-B and association between CH1-B and CL-A are inhibited because the amino acid residues forming the interfaces mutually have the same charge. As a result, antibodies having the intended association between CH1-A and CL-A, and the intended association between CH1-B and CL-B can be acquired efficiently. Moreover, the intended association between CH1-A and CL-A is promoted since the amino acid residues forming the interface have different types of charges from each other; and the intended association between CH1-B and CL-B is also promoted since the amino acid residues forming the interface have different types of charges from each other. Consequently, antibodies with intended association can be efficiently obtained.

Another example is modifying the amino acid residues forming the interface between CH1-A and CL-B into positively charged amino acid residues, when the amino acid residues forming the interface between CL-A and CH1-B are mutually uncharged or nonpolar amino acids. As a result of this modification, the unintended association between CH1-A and CL-B is inhibited because the amino acid residues forming the interface are both positively charged. On the other hand, since the amino acid residues forming the interfaces are amino acids that do not mutually repel electrically, the intended association between CH1-A and CL-A, and the intended association between CH1-B and CL-B will occur more easily than in the case where the amino acids repel electrically. Consequently, antibodies having the intended association between CH1-A and CL-A, and the intended association between CH1-B and CL-B can be efficiently obtained. Meanwhile, in this example, in the case that the amino acid residues forming the interface between CL-A and CH1-B are not mutually uncharged or nonpolar amino acids, they may be modified so as to become mutually uncharged or nonpolar amino acids.

In another example, when amino acid residues forming the interface between CL-B and CH1-B are mutually uncharged or nonpolar amino acids, one of the amino acid residues forming the interface between CH1-A and CL-A is modified into a positively charged amino acid residue, and the other is modified into a negatively charged amino acid residue. As a result of this modification, while the intended association between CH1-A and CL-A is promoted because the amino acid residues forming the interface are a combination of positive charge and negative charge, the intended association between CH1-B and CL-B is not inhibited because the amino acid residues forming the interface are amino acids that do not mutually repel electrically. As a result, one can efficiently obtain an antibody having intended association between CH1-A and CL-A, and intended association between CH1-B and CL-B. Meanwhile, in this example, when the amino acid residues forming the interface between CL-B and CH1-B are not mutually uncharged or nonpolar amino acids, they may be modified so as to become mutually uncharged or nonpolar amino acids.

Moreover, in another example, when the amino acid residues forming the interface between CL-B and CH1-B are uncharged or nonpolar amino acids in CH1-B, one of the amino acid residues forming the interface between CH1-A and CL-A is modified into a positively charged amino acid residue while the other is modified into a negatively charged amino acid residue; and amino acid residues forming the interface between CL-B and CH1-B in CL-B are modified so as to have the same charge as the modification made to CH1-A. As a result of this modification, while the intended association between CH1-A and CL-A is promoted because the amino acid residues forming the interface are a combination of positive charge and negative charge, the intended association between CH1-B and CL-B is not inhibited because the amino acid residues forming the interface are amino acids that do not mutually repel electrically. As a result, one can efficiently obtain an antibody having intended association between CH1-A and CL-A, and intended association between CH1-B and CL-B. Meanwhile, in this example, when the amino acid residues forming the interface between CL-B and CH1-B are not uncharged or nonpolar amino acids in CH1-B, they may be modified so as to become uncharged or nonpolar amino acids.

In addition, use of the association regulation of the present invention makes it possible to suppress association between CHls (CH1-A and CH1-B), or association between CLs (CL-A and CL-B).

Those skilled in the art would be able to suitably determine the types of amino acid residues that come close during association at the CH1 and CL interface in a desired polypeptide for which regulation of association by the present invention is desired.

Further, those skilled in the art can also suitably acquire sequences that can be used as CH1 or CL of an antibody in an organism such as a human, monkey, mouse, rabbit, and the like by using a public database and such. More specifically, the amino acid sequence information of CH1 or CL can be acquired by means described in the Examples described below.

For example, with respect to the bispecific antibodies described in the Examples below, specific examples of amino acid residues that come close (that face or are in contact) at the interface of CH1 and CL upon association include the combinations shown below:

    • lysine (K) at position 147 as indicated by EU numbering in CH1 (for example, position 147 in the amino acid sequence of SEQ ID NO: 1) and the facing (contacting) threonine (T) at position 180 as indicated by EU numbering in CL;
    • lysine (K) at position 147 as indicated by EU numbering in CH1 and the facing (contacting) serine (S) at position 131 as indicated by EU numbering in CL;
    • glutamine (Q) at position 175 as indicated by EU numbering in CH1 and the facing (contacting) glutamine (Q) at position 160 as indicated by EU numbering in CL; and,
    • lysine (K) at position 213 as indicated by EU numbering in CH1 and the facing (contacting) glutamic acid (E) at position 123 as indicated by EU numbering in CL.

The numbers described in EU numbering in the present invention are indicated in accordance with EU numbering (Sequences of proteins of immunological interest, NIH Publication No. 91-3242). In the present invention, the phrases “an amino acid residue at position X as indicated by EU numbering” and “an amino acid at position X as indicated by EU numbering” (where X is an arbitrary number) can also be read as “an amino acid residue that corresponds to position X as indicated by EU numbering” and “an amino acid that corresponds to position X as indicated by EU numbering”.

As indicated in the Examples described below, desired antibodies can be preferentially acquired by modifying these amino acid residues and carrying out the methods of the present invention.

Accordingly, the present invention provides an antibody in which association of the heavy chain and light chain is regulated, wherein one or two or more sets of amino acid residues selected from the group consisting of the sets of amino acid residues shown in (a) to (c) below in the heavy chain and light chain of the antibody are amino acid residues that mutually repel electrically:

(a) the amino acid residue contained in the heavy chain constant region (CH1) at position 147 as indicated by EU numbering, and the amino acid residue contained in the light chain constant region (CL) at position 180 as indicated by EU numbering;
(b) the amino acid residue contained in CH1 at position 147 as indicated by EU numbering, and the amino acid residue contained in CL at position 131 as indicated by EU numbering; and
(c) the amino acid residue contained in CH1 at position 175 as indicated by EU numbering, and the amino acid residue contained in CL at position 160 as indicated by EU numbering.

As another embodiment, the present invention further provides an antibody in which the amino acid residues in the set of the amino acid residues of (d) below are amino acid residues that mutually repel electrically:

(d) the amino acid residue contained in CH1 at position 213 as indicated by EU numbering, and the amino acid residue contained in CL at position 123 as indicated by EU numbering.

In the aforementioned antibody, the “amino acid residues that mutually repel electrically” or “amino acid residues having the same charge” are preferably selected from amino acid residues contained in, for example, either of the set of (X) or (Y) below:

(X) glutamic acid (E) or aspartic acid (D); or
(Y) lysine (K), arginine (R), or histidine (H).

In the aforementioned antibody, specific examples of amino acid residues that mutually repel electrically include the amino acid residues below:

    • the amino acid residue contained in CH1 at position 175 as indicated by EU numbering is lysine (K), and the amino acid residues contained in CL at position 180, position 131, and position 160 as indicated by EU numbering are all glutamic acid (E); and,
    • the amino acid residues contained in CH1 at position 147 and position 175 as indicated by EU numbering are glutamic acid (E), and the amino acid residues contained in CL at position 180, position 131, and position 160 as indicated by EU numbering are all lysine (K).

In the aforementioned antibody, examples of amino acid residues that do not electrically repel further include one in which the amino acid residue contained in CH1 at position 213 as indicated by EU numbering is glutamic acid (E), and the amino acid residue contained in CL at position 123 as indicated by EU numbering is lysine (K).

Moreover, methods for producing an aforementioned antibody and methods of the present invention for regulating association through modification of the amino acid residues in the sets of amino acid residues of (a) to (d) mentioned above into amino acid residues that mutually repel electrically are also preferred embodiments of the present invention.

Further, the present invention provides an antibody in which association of the heavy chain and light chain is regulated, wherein one or two or more sets of amino acid residues selected from the group consisting of the sets of amino acid residues shown in (a) to (c) below in the associating heavy chain and light chain of the antibody are amino acid residues that do not mutually repel electrically:

(a) the amino acid residue contained in CH1 at position 147 as indicated by EU numbering, and the amino acid residue contained in CL at position 180 as indicated by EU numbering;
(b) the amino acid residue contained in CH1 at position 147 as indicated by EU numbering, and the amino acid residue contained in CL at position 131 as indicated by EU numbering; and
(c) the amino acid residue contained in CH1 at position 175 as indicated by EU numbering, and the amino acid residue contained in CL at position 160 as indicated by EU numbering.

As another embodiment, the present invention further provides an antibody in which the amino acid residues in the set of amino acid residues shown in (d) below are amino acid residues that do not mutually repel electrically:

(d) the amino acid residue contained in CH1 at position 213 as indicated by EU numbering, and the amino acid residue contained in CL at position 123 as indicated by EU numbering.

As indicated in the Examples described below and FIG. 1, each of the amino acid residues of the aforementioned combinations mutually approaches upon association. Those skilled in the art would be able to find sites corresponding to the amino acid residues described in (a) to (d) mentioned above for the desired CH1 or CL by homology modeling and such using commercially available software, and to suitably modify the amino acid residues at those sites.

In the aforementioned antibody, the “amino acid residues that do not mutually repel electrically” are preferably selected from, for example, each of the two sets selected from the group consisting of (X) to (Z) shown below, and where the two sets are selected from among the combinations of (X) and (Y), (X) and (Z), (Y) and (Z), and (Z) and (Z):

(X) glutamic acid (E) or aspartic acid (D);
(Y) lysine (K), arginine (R) or histidine (H);
(Z) alanine (A), asparagine (N), cysteine (C), glutamine (Q), glycine (G), isoleucine (I), leucine (L), methionine (M), phenylalanine (F), proline (P), serine (S), threonine (T), tryptophan (W), tyrosine (Y) or valine (V).

An example involves selecting (X) and (Y) from the group consisting of (X) to (Z), selecting glutamic acid from (X) and selecting lysine (K) from (Y); and modifying the amino acid residue contained in CH1 at position 147 as indicated by EU numbering to glutamic acid (E), and modifying the amino acid residue contained in CL at position 180 as indicated by EU numbering to lysine (K). In this case, there is no need to modify the amino acid residue contained in CH1 at position 147 as indicated by EU numbering if the amino acid residue is glutamic acid (E) from before the modification.

In the aforementioned antibody, specific examples of amino acid residues that do not mutually repel electrically include the amino acid residues shown below:

    • the amino acid residue contained in CH1 at position 175 as indicated by EU numbering is lysine (K), and the amino acid residues contained in CL at position 180, position 131, and position 160 as indicated by EU numbering are all glutamic acid (E); and
    • the amino acid residues contained in CH1 at position 147 and position 175 as indicated by EU numbering are glutamic acid (E), and the amino acid residues contained in CL at position 180, position 131, and position 160 as indicated by EU numbering are all lysine (K).

In the aforementioned antibody, examples of amino acid residues that do not electrically repel further include one in which the amino acid residue contained in CH1 at position 213 as indicated by EU numbering is glutamic acid (E), and the amino acid residue contained in CL at position 123 as indicated by EU numbering is lysine (K).

Moreover, methods for producing an aforementioned antibody, and methods of the present invention for regulating association through modification of the amino acid residues in the sets of amino acid residues in (a) to (d) mentioned above into amino acid residues that do not mutually repel electrically are also preferred embodiments of the present invention.

A technique for introducing electrical repulsion into the interface of the second constant region of the heavy chain (CH2) or the third constant region of the heavy chain (CH3) to suppress undesired association between heavy chains, a technique for introducing electrical repulsion into the interface of the heavy chain variable region and light chain variable region to suppress unintended association between the heavy chain and light chain, or a technique for modifying amino acid residues forming a hydrophobic core present at the interface of the heavy chain variable region and light chain variable region into polar amino acids having an electrical charge to suppress unintended association between the heavy chain and light chain can be further applied to the antibody of the present invention (see WO 2006/106905).

In the technique that suppresses unintended association between heavy chains by introducing electrical repulsion at the interface of CH2 or CH3, examples of amino acid residues that are in contact at the interface of other constant regions of the heavy chain include regions corresponding to position 377 (position 356) and position 470 (position 439), position 378 (position 357) and position 393 (position 370), and position 427 (position 399) and position 440 (position 409) in the CH3 region. For the numbering of the antibody constant regions, one may refer to the publication by Kabat et al. (Kabat, E. A., et al., 1991, Sequences of Proteins of Immunological Interest, NIH); and for the numbering of the heavy chain constant regions, the EU numbering are shown inside the parentheses.

The technique of modifying the amino acid residue at position 435 as indicated by EU numbering, which is a site related to binding between IgG and Protein A, to an amino acid having a different binding strength toward Protein A, such as Arg, may also be used on the antibody of the present invention in combination with the aforementioned techniques. By using this technique, the interaction between the H chain and Protein A can be changed, and only heterodimeric antibodies can be efficiently purified using a Protein A column. This technique can also be used independently without combining with the aforementioned techniques.

More specifically, for example, in an antibody containing two types of heavy chain CH3 regions, one to three sets of amino acid residues in the first heavy chain CH3 region, which are selected from the sets of amino acid residues of (1) to (3) below, may be made to mutually repel electrically:

(1) the amino acid residues contained in the heavy chain CH3 region at position 356 and position 439 as indicated by EU numbering;
(2) the amino acid residues contained in the heavy chain CH3 region at position 357 and position 370 as indicated by EU numbering; and
(3) the amino acid residues contained in the heavy chain CH3 region at position 399 and position 409 as indicated by EU numbering.

Moreover, the antibody can be an antibody having a set of amino acid residues in the second heavy chain CH3 region distinct from the aforementioned first heavy chain CH3 region, wherein the set of amino acid residues is selected from the sets of amino acid residues shown in (1) to (3) above, and wherein the one to three sets of amino acid residues that correspond to the sets of amino acid residues shown in (1) to (3) above, which mutually repel electrically in the first heavy chain CH3 region, do not electrically repel from the corresponding amino acid residues in the first heavy chain CH3 region.

The amino acid residues described in (1) to (3) above approach each other upon association. Those skilled in the art would be able to find sites corresponding to the amino acid residues described in (1) to (3) mentioned above for a desired heavy chain CH3 region or heavy chain constant region by homology modeling and such using commercially available software, and to suitably modify the amino acid residues at those sites.

In the aforementioned antibody, “electrically repelling” or “having a same charge” means that, for example, any two or more amino acid residues have amino acid residues that are contained in either one group of (X) and (Y) mentioned above. On the other hand, “not electrically repelling” means that, for example, the antibody has amino acid residues that are selected from each of two sets selected from the group consisting of (X) and (Y) mentioned above and (Z) below, where the two sets are selected from among the combinations of (X) and (Y), (X) and (Z), (Y) and (Z), and (Z) and (Z):

(Z) alanine (A), asparagine (N), cysteine (C), glutamine (Q), glycine (G), isoleucine (I), leucine (L), methionine (M), phenylalanine (F), proline (P), serine (S), threonine (T), tryptophan (W), tyrosine (Y) or valine (V).

In a preferred embodiment of the aforementioned antibody, the first heavy chain CH3 region and the second heavy chain CH3 region may be cross-linked by disulfide bonds.

In the present invention, an amino acid residue subjected to “modification” is not limited to an amino acid residue of the antibody variable region or antibody constant region mentioned above. Those skilled in the art would be able to find amino acid residues that form an interface in a polypeptide variant or heteromeric multimer by homology modeling and the like using commercially available software, and to modify amino acid residues at those sites so as to regulate association. Homology modeling is a technique for predicting the three-dimensional structure of a protein using commercially available software. When constructing the structure of a protein with unknown three-dimensional structure, one first searches for a protein that has been determined to have a highly homologous three-dimensional structure to the protein. Next, using this three-dimensional structure as a template, one constructs the structure of the protein with unknown structure, and the structure is further optimized by molecular dynamics methods and the like to predict the three-dimensional structure of the unknown protein.

In the technique for introducing electrical repulsion into the interface of the heavy chain variable region and light chain variable region to suppress undesired association of the heavy chain and light chain, examples of amino acid residues that are in contact at the interface of the heavy chain variable region (VH) and light chain variable region (VL) include glutamine (Q) at position 39 as indicated by Kabat numbering in the heavy chain variable region (FR2 region) and the facing (contacting) glutamine (Q) at position 38 as indicated by Kabat numbering in the light chain variable region (FR2 region). Moreover, a preferable example is leucine (L) at position 45 according to the Kabat numbering in the heavy chain variable region (FR2) and the facing proline (P) at position 44 according to the Kabat numbering in the light chain variable region (FR2). The publication by Kabat, et al. (Kabat, E. A., et al., 1991, Sequence of Proteins of Immunological Interest, NIH) was referred to for the numbering of these sites.

Since these amino acid residues are known to be highly conserved in humans and mice (J. Mol. Recognit. 2003; 16: 113-120), association of antibody variable regions can be regulated for VH-VL association of antibodies other than those indicated in the Examples by modifying amino acid residues corresponding to the above-mentioned amino acid residues.

A specific example is an antibody in which two or more amino acid residues forming the interface of the heavy chain variable region and light chain variable region are amino acid residues that mutually repel electrically.

More specifically, examples include an antibody with one set or two sets of amino acid residues selected from the group consisting of the sets of amino acid residues shown in (a) or (b) below:

(a) the amino acid residue contained in the heavy chain variable region (1) at position 39 as indicated by Kabat numbering and the amino acid residue contained in the light chain (2) at position 38 as indicated by Kabat numbering; or
(b) the amino acid residue contained in the heavy chain variable region (3) at position 45 as indicated by Kabat numbering and the amino acid residue contained in the light chain variable region (4) at position 44 as indicated by Kabat numbering.

Each of the amino acid residues described in the aforementioned (a) or (b) approaches each other upon association. Those skilled in the art would be able to find sites that correspond to the amino acid residues described in the aforementioned (a) or (b) in a desired heavy chain variable region or light chain variable region by homology modeling and the like using commercially available software, and to suitably modify the amino acid residues at those sites.

In the aforementioned antibody, “amino acid residues that mutually repel electrically” are preferably selected from amino acid residues contained in, for example, either of the sets (X) and (Y) below:

(X) glutamic acid (E) or aspartic acid (D); or
(Y) lysine (K), arginine (R), or histidine (H).

In addition, another embodiment of the antibody of the present invention is, for example, an antibody in which two or more amino acid residues that form the interface of the heavy chain variable region and light chain variable region are amino acid residues that do not electrically repel each other. Specifically, an example of such an antibody is one having one set or two sets of amino acid residues selected from the group consisting of the sets of amino acid residues shown in the aforementioned (a) and (b).

The respective amino acid residues described in the aforementioned (a) or (b) are close to each other upon association. Those skilled in the art would be able to find sites that correspond to the amino acid residues described in the aforementioned (a) or (b) for a desired heavy chain variable region or light chain variable region by homology modeling and the like using commercially available software, and to suitably subject amino acid residues at those sites to modification.

In the aforementioned antibody, “amino acid residues that do not mutually repel electrically” refers to, for example, amino acid residues selected from each of two sets selected from the group consisting of (X) to (Z) mentioned above, and where the two sets are selected from among the combinations of (X) and (Y), (X) and (Z), (Y) and (Z), and (Z) and (Z).

Generally, the amino acid residues described in the aforementioned (a) or (b) in humans and mice are:

(1) glutamine (Q),
(2) glutamine (Q),
(3) leucine (L), and
(4) proline (P).
Thus, in a preferred embodiment of the present invention, these amino acid residues are subjected to modification (such as substitution with amino acids having an electrical charge). Furthermore, the types of amino acid residues of the aforementioned (a) or (b) are not necessarily limited to the aforementioned amino acid residues, but may also be other amino acids equivalent to these amino acids. For example, the amino acid at position 38 as indicated by Kabat numbering in the light chain variable region may be, for example, histidine (H) in the case of humans.

In the technique for modifying amino acid residues forming a hydrophobic core present at the interface of the heavy chain variable region and light chain variable region into polar amino acids having an electrical charge to suppress unintended association of the heavy chain and light chain, preferable examples of amino acid residues which are able to form a hydrophobic core at the interface of the heavy chain variable region (VH) and light chain variable region (VL) include leucine (L) at position 45 as indicated by Kabat numbering in the heavy chain variable region (FR2) and the facing proline (P) at position 44 as indicated by Kabat numbering in light chain variable region (FR2). For the numbering of these sites, Kabat, et al. (Kabat, E. A., et al., 1991, Sequences of Proteins of Immunological Interest, NIH) was used as a reference.

In general, the term “hydrophobic core” refers to a part that is formed by an assembly of hydrophobic amino acid side chains at the interior of associated polypeptides. Examples of hydrophobic amino acids include alanine, isoleucine, leucine, methionine, phenylalanine, proline, tryptophan, and valine. Furthermore, amino acid residues other than hydrophobic amino acids (for example tyrosine) may be involved in the formation of a hydrophobic core. This hydrophobic core together with a hydrophilic surface, in which hydrophilic amino acid side chains are exposed to the exterior, becomes a driving force for promoting association of water-soluble polypeptides. When hydrophobic amino acids of two different domains are present on a molecular surface and are exposed to water molecules, the entropy will increase and the free energy will increase. Accordingly, the two domains will associate with each other to decrease the free energy and become stable, and hydrophobic amino acids at the interface will be buried into the interior of the molecule to form a hydrophobic core.

It is thought that when polypeptide association occurs, formation of a hydrophobic core is inhibited by modifying hydrophobic amino acids forming the hydrophobic core to polar amino acids having an electrical charge; and consequently, polypeptide association is thought to be inhibited.

Those skilled in the art would be able to recognize the presence or absence of a hydrophobic core, the formation site (region), and the like by analyzing amino acid sequences for a desired polypeptide. Namely, the antibody of the present invention is an antibody characterized in that amino acid residues capable of forming a hydrophobic core at an interface are modified to amino acid residues having an electrical charge. More specifically, examples include an antibody in which the amino acid residues shown in either (1) or (2) below are amino acid residues having an electrical charge. Side chains of the amino acid residues shown in (1) and (2) below are adjacent to each other, and can form a hydrophobic core:

(1) the amino acid residue contained in the heavy chain variable region at position 45 as indicated by Kabat numbering; and
(2) the amino acid residue contained in the light chain variable region at position 44 as indicated by Kabat numbering.

Preferable examples of amino acid residues having an electrical charge in the aforementioned antibody include glutamic acid (E), aspartic acid (D), lysine (K), arginine (R) and histidine (H). More preferable examples include glutamic acid (E) and lysine (K).

Generally, the amino acid residues described in the aforementioned (1) and (2) in humans and mice are respectively:

(1) leucine (L), and
(2) proline (P).
Thus, in a preferred embodiment of the present invention, these amino acid residues are subjected to modification (such as substitution with amino acids having an electrical charge). Furthermore, the types of the aforementioned amino acid residues of (1) and (2) are not necessarily limited to the aforementioned amino acid residues, but may also be other amino acids equivalent to these amino acid residues.

Other known techniques can be applied to the antibodies of the present invention. For example, in order to promote association of the first VH (VH1) and the first VL (VL1) and/or the second VH (VH2) and the second VL (VL2), an amino acid side chain present in the variable region of one of the H chains can be substituted with a larger side chain (knob), and an amino acid side chain present in the opposing variable region of the other H chain can be substituted with a smaller side chain (hole), so that the knob may be arranged in the hole, and association of VH1 and VL1 and/or VH2 and VL2 is promoted; and consequently, association of VH1 and VL2 and/or VH2 and VL1 can be further suppressed (WO 1996/027011; Ridgway, J. B., et al., Protein Engineering (1996) 9, 617-621; Merchant, A. M., et al., Nature Biotechnology (1998) 16, 677-681).

For example, in the case of human IgG1, in order to make an amino acid side chain in the CH3 region of one H chain a larger side chain (knob), the modifications of Y349C and T366W are made, and in order to make an amino acid side chain in the CH3 region of the other H chain a smaller side chain, the modifications of D356C, T336S, L368A and Y407V are made.

Still other known techniques can be applied to the antibodies of the present invention. A target antibody can be efficiently prepared by complementary association of CH3 using strand-exchange engineered domain CH3, in which a portion of CH3 of one H chain of an antibody is changed to a sequence derived from IgA corresponding to that portion, and a complementary portion of CH3 of the other H chain is introduced with a sequence derived from IgA corresponding to that portion (Protein Engineering Design & Selection, 23: 195-202, 2010).

Still other known techniques can be applied to the antibodies of the present invention. When producing bispecific antibodies, a target bispecific antibody can be prepared by, for example, imparting a difference in isoelectric point by making different amino acid modifications to each of the variable regions of the two types of H chains, and utilizing that difference in isoelectric point for purification by ion exchange chromatography (WO 2007/114325).

The modifications of the present invention can be used on antibodies such as the one below, for example, an antibody having a structure in which, to promote association of a first VH

(VH1) and a first VL (VL1) and/or a second VH (VH2) and a second VL (VL2), VH1 is linked to an Fc region through a first CH1 and VL1 is linked to a first CL, and VH2 is linked to another Fc region through a second CL and VL2 is linked to a second CH1 (WO 09/80254).

A plurality, for example, two or more of the aforementioned known techniques can be used in combination for the antibody of the present invention. Furthermore, the antibody of the present invention may be prepared based on an antibody to which modifications of the aforementioned known techniques have been made.

The below-mentioned methods of the present invention for regulating association allow, for example, for the efficient production of antibodies or polypeptides that are active. Examples of such activities include binding activity, neutralizing activity, cytotoxic activity, agonist activity, antagonist activity, and enzyme activity and such. Agonist activity is an activity that induces some kind of changes in physiological activity through binding of an antibody to an antigen, such as a receptor, which causes signal transduction or such in cells. Examples of the physiological activity include growth activity, survival activity, differentiation activity, transcriptional activity, membrane transport activity, binding activity, proteolytic activity, phosphorylation/dephosphorylation activity, redox activity, transfer activity, nucleolytic activity, dehydration activity, cell death-inducing activity, and apoptosis-inducing activity and such, but are not limited thereto.

Antibodies or polypeptides that recognize the desired antigens or bind to the desired receptors can be produced efficiently by the methods of the present invention.

The antigens of the present invention are not particularly limited, and any type of antigen can be used. Examples of antigens include receptors or their fragments, cancer antigens, MHC antigens, and differentiation antigens and the like, but are not particularly limited thereto.

Examples of the receptors of the present invention include receptors belonging to the hematopoietic factor receptor family, cytokine receptor family, tyrosine kinase-type receptor family, serine/threonine kinase-type receptor family, TNF receptor family, G protein-coupled receptor family, GPI-anchored receptor family, tyrosine phosphatase-type receptor family, adhesion factor family, hormone receptor family, and such. Reports on the receptors belonging to these receptor families and their characteristics can be found in various sources of documents, for example, in Cooke B A., King R J B., van der Molen H J. ed. New Comprehensive Biochemistry Vol. 18B “Hormones and their Actions Part II” pp. 1-46 (1988) Elsevier Science Publishers BV., New York, USA; Patthy L. (1990) Cell, 61: 13-14; Ullrich A., et al. (1990) Cell, 61: 203-212; Massagul J. (1992) Cell, 69: 1067-1070; Miyajima A., et al. (1992) Annu. Rev. Immunol., 10: 295-331; Taga T. and Kishimoto T. (1992) FASEB J., 7: 3387-3396; Fantl W I., et al. (1993) Annu. Rev. Biochem., 62: 453-481; Smith C A., et al. (1994) Cell, 76: 959-962; Flower D R. (1999) Biochim. Biophys. Acta, 1422: 207-234; Miyasaka M. ed. Cell Technology, Handbook Series “Handbook for adhesion factors” (1994) Shujunsha, Tokyo, Japan; and such. Examples of specific receptors belonging to the above-mentioned receptor families include human or mouse erythropoietin (EPO) receptor, human or mouse granulocyte-colony stimulating factor (G-CSF) receptor, human or mouse thrombopoietin (TPO) receptor, human or mouse insulin receptor, human or mouse Flt-3 ligand receptor, human or mouse platelet-derived growth factor (PDGF) receptor, human or mouse interferon (IFN)-α or -βreceptor, human or mouse leptin receptor, human or mouse growth hormone (GH) receptor, human or mouse interleukin (IL)-10 receptor, human or mouse insulin-like growth factor (IGF)-I receptor, human or mouse leukemia inhibitory factor (LIF) receptor, and human or mouse ciliary neurotrophic factor (CNTF) receptor (hEPOR: Simon, S. et al. (1990) Blood 76, 31-35; mEPOR: D'Andrea, A D. et al. (1989) Cell 57, 277-285; hG-CSFR: Fukunaga, R. et al. (1990) Proc. Natl. Acad. Sci. USA.

87, 8702-8706; mG-CSFR: Fukunaga, R. et al. (1990) Cell 61, 341-350; hTPOR: Vigon, I. et al. (1992) 89, 5640-5644.; mTPOR: Skoda, R C. et al. (1993) 12, 2645-2653; hInsR: Ullrich, A. et al. (1985) Nature 313, 756-761; hFlt-3: Small, D. et al. (1994) Proc. Natl. Acad. Sci. USA. 91, 459-463; hPDGFR: Gronwald, R G K. et al. (1988) Proc. Natl. Acad. Sci. USA. 85, 3435-3439; hIFN α/β R: Uze, G. et al. (1990) Cell 60, 225-234; and Novick, D. et al. (1994) Cell 77, 391-400).

Cancer antigens are antigens that are expressed following malignant transformation of a cell, and are also referred to as tumor specific antigens. In addition, abnormal sugar chains which appear on a cell surface or on a protein molecule when the cell has become cancerous are also cancer antigens, and they are also referred to as cancer sugar chain antigens. Examples of cancer antigens include EpCAM, which is expressed in multiple cancers including lung cancer (Proc. Natl. Acad. Sci. USA (1989) 86 (1), 27-31) (the polynucleotide sequence thereof is indicated as RefSeq Accession No. NM_002354.2 (SEQ ID NO: 78) and the polypeptide sequence thereof is indicated as RefSeq Accession No. NP_002345.2 (SEQ ID NO: 79)), CA19-9, CA15-3, sialyl SSEA-1 (SLX), etc.

MHC antigens can be classified broadly into MHC class I antigens and MHC class II antigens: MHC class I antigens include HLA-A, -B, -C, -E, -F, -G, and -H; and MHC class II antigens include HLA-DR, -DQ, and -DP.

Differentiation antigens include CD1, CD2, CD3, CD4, CD5, CD6, CD7, CD8, CD10, CD11a, CD11b, CD11c, CD13, CD14, CD15s, CD16, CD18, CD19, CD20, CD21, CD23, CD25, CD28, CD29, CD30, CD32, CD33, CD34, CD35, CD38, CD40, CD41a, CD41b, CD42a, CD42b, CD43, CD44, CD45, CD45RO, CD48, CD49a, CD49b, CD49c, CD49d, CD49e, CD49f, CD51, CD54, CD55, CD56, CD57, CD58, CD61, CD62E, CD62L, CD62P, CD64, CD69, CD71, CD73, CD95, CD102, CD106, CD122, CD126, and CDw130.

The antibodies of the present invention may be a bispecific antibody; and in that case, two antigens (or epitopes) recognized by that antibody can be arbitrarily selected from the aforementioned receptors or fragments thereof, cancer antigens, MHC antigens, differentiation antigens and the like. For example, two antigens may be selected from receptors or fragments thereof, two may be selected from cancer antigens, two may be selected from MHC antigens, or two may be selected from differentiation antigens. In addition, one antigen each may be selected from two antigens arbitrarily selected from, for example, receptors or fragments thereof, cancer antigens, MHC antigens, and differentiation antigens.

In addition, the present invention provides a method for producing an antibody in which association of the heavy chain and light chain is regulated.

A preferred embodiment of the production method of the present invention is a method for producing an antibody in which association of the heavy chain and light chain is regulated, comprising:

(1) modifying nucleic acids encoding CH1 and CL so that one set or two or more sets of amino acid residues selected from the group consisting of the sets of amino acid residues shown in (a) to (c) below become amino acid residues that electrically repel each other:
(a) the amino acid residue contained in CH1 at position 147 as indicated by EU numbering, and the amino acid residue contained in CL at position 180 as indicated by EU numbering;
(b) the amino acid residue contained in CH1 at position 147 as indicated by EU numbering, and the amino acid residue contained in CL at position 131 as indicated by EU numbering; and
(c) the amino acid residue contained in CH1 at position 175 as indicated by EU numbering, and amino acid residue contained in CL at position 160 as indicated by EU numbering,
(2) introducing the modified nucleic acids into host cells and culturing so that the host cells express the nucleic acids, and
(3) collecting an antibody from a cell culture of the host cells.

Another embodiment of the production method of the present invention includes a method for producing an antibody, wherein step (1) of the aforementioned production method further comprises modifying the nucleic acids so that the amino acid residues of the set of amino acid residues shown in (d) below become amino acid residues that electrically repel each other:

(d) the amino acid residue contained in CH1 at position 213 as indicated by EU numbering, and the amino acid residue contained in CL at position 123 as indicated by EU numbering.

In addition, the present invention relates to a production method comprising, in the aforementioned step (1), modifying the nucleic acids so that the amino acid residues that electrically repel each other are selected from among the amino acid residues contained in either of the groups of the aforementioned (X) and (Y).

Moreover, the present invention relates to a production method comprising in the aforementioned step (1), modifying the nucleic acids so that two or more amino acid residues that form the interface of the heavy chain variable region and light chain variable region are amino acid residues that electrically repel each other. Preferably, the amino acid residues that electrically repel each other are any set of amino acid residues selected from the group consisting of, for example, the sets of amino acid residues shown in (a) and (b) below:

(a) the amino acid residue contained in the heavy chain variable region at position 39 as indicated by Kabat numbering, and the amino acid residue contained in the light chain variable region at position 38 as indicated by Kabat numbering; or
(b) the amino acid residue contained in the heavy chain variable region at position 45 as indicated by Kabat numbering, and the amino acid residue contained in the light chain variable region at position 44 as indicated by Kabat numbering.

The aforementioned amino acid residues which electrically repel each other are preferably selected from the amino acid residues contained in either set of the aforementioned (X) and (Y).

Another preferred embodiment of the production method of the present invention includes a method for producing an antibody in which association of the heavy chain and light chain is regulated, comprising:

(1) modifying nucleic acids encoding CH1 and CL so that one set or two or more sets of amino acid residues selected from the group consisting of the sets of amino acid residues shown in (a) to (c) below become amino acid residues that do not electrically repel each other:
(a) the amino acid residue contained in CH1 at position 147 as indicated by EU numbering, and the amino acid residue contained in CL at position 180 as indicated by EU numbering;
(b) the amino acid residue contained in CH1 at position 147 as indicated by EU numbering, and the amino acid residue contained in CL at position 131 as indicated by EU numbering; and
(c) the amino acid residue contained in CH1 at position 175 as indicated by EU numbering, and the amino acid residue contained in CL at position 160 as indicated by EU numbering,
(2) introducing the modified nucleic acids into host cells and culturing so that the host cells express the nucleic acids, and
(3) collecting an antibody from a culture of the host cells.

Another embodiment of the production method of the present invention includes a production method further comprising in step (1) of the aforementioned production method, modifying the nucleic acids so that the amino acid residues of the set of amino acid residues shown in (d) below become amino acid residues that do not electrically repel each other:

(d) the amino acid residue contained in CH1 at position 213 as indicated by EU numbering, and the amino acid residue contained in CL at position 123 as indicated by EU numbering.

In addition, the present invention relates to a production method comprising, in the aforementioned step (1), modifying nucleic acids so that the amino acid residues that do not electrically repel each other are amino acid residues selected from each of two sets selected from the group consisting of the aforementioned (X) to (Z), and where the two sets are selected from among the combinations of (X) and (Y), (X) and (Z), (Y) and (Z), and (Z) and (Z).

In addition, specific examples of the amino acid residues that do not electrically repel each other in the aforementioned step (1) of the present invention include the following amino acid residues:

    • the amino acid residue contained in CH1 at position 175 as indicated by EU numbering which is lysine (K); and the amino acid residues contained in CL at position 180, position 131, and position 160 as indicated by EU numbering which are all glutamic acid (E); and
    • the amino acid residues contained in CH1 at position 147 and position 175 as indicated by EU numbering which are glutamic acid (E), and the amino acid residues contained in CL at position 180, position 131, and position 160 as indicated by EU numbering which are all lysine (K).

Moreover, in another example, the amino acid residue contained in CH1 at position 213 as indicated by EU numbering is glutamic acid (E), and the amino acid residue contained in CL at position 123 as indicated by EU numbering is lysine (K).

Moreover, the present invention relates to a production method, comprising in the aforementioned step (1), modifying the nucleic acids so that two or more amino acid residues that form the interface of the heavy chain variable region and light chain variable region are amino acid residues that do not electrically repel each other. Preferably, the amino acid residues that do not electrically repel each other are, for example, amino acid residues of any set selected from the group consisting of the sets of amino acid residues indicated in (a) and (b) below:

(a) the amino acid residue contained in the heavy chain variable region at position 39 as indicated by Kabat numbering, and the amino acid residue contained in the light chain variable region at position 38 as indicated by Kabat numbering; and
(b) the amino acid residue contained in the heavy chain variable region at position 45 as indicated by Kabat numbering, and the amino acid residue contained in the light chain variable region at position 44 as indicated by Kabat numbering.

The aforementioned amino acid residues that do not electrically repel each other are preferably amino acid residues selected from each of two sets selected from the group consisting of the aforementioned (X) to (Z), and where the two sets are selected from among the combinations of (X) and (Y), (X) and (Z), (Y) and (Z), and (Z) and (Z).

In addition, the present invention provides a method for regulating association of the heavy chains and light chains of an antibody.

A preferred embodiment of the method for regulating association of the present invention is a method for regulating association of the heavy chains and light chains of an antibody, comprising modifying nucleic acids so that one set or two or more sets of amino acid residues selected from the group consisting of the sets of amino acid residues shown in (a) to (c) below become amino acid residues that electrically repel each other:

(a) the amino acid residue contained in CH1 at position 147 as indicated by EU numbering, and the amino acid residue contained in CL at position 180 as indicated by EU numbering;
(b) the amino acid residue contained in CH1 at position 147 as indicated by EU numbering, and the amino acid residue contained in CL at position 131 as indicated by EU numbering; and
(c) the amino acid residue contained in CH1 at position 175 as indicated by EU numbering, and the amino acid residue contained in CL at position 160 as indicated by EU numbering.

Another embodiment of the present invention provides a method for regulating association in an antibody, further comprising modifying nucleic acids so that amino acid residues of the set of amino acid residues shown in (d) below are amino acid residues that electrically repel each other:

(d) the amino acid residue contained in CH1 at position 213 as indicated by EU numbering, and the amino acid residue contained in CL at position 123 as indicated by EU numbering.

Another preferred embodiment of the method for regulating association of the present invention is a method for regulating association of the heavy chains and light chains of an antibody, comprising modifying nucleic acids so that one set or two or more sets of amino acid residues selected from the group consisting of the sets of amino acid residues shown in (a) to (c) below become amino acid residues that do not electrically repel each other:

(a) the amino acid residue contained in CH1 at position 147 as indicated by EU numbering, and the amino acid residue contained in CL at position 180 as indicated by EU numbering;
(b) the amino acid residue contained in CH1 at position 147 as indicated by EU numbering, and the amino acid residue contained in CL at position 131 as indicated by EU numbering; and
(c) the amino acid residue contained in CH1 at position 175 as indicated by EU numbering, and the amino acid residue contained in CL at position 160 as indicated by EU numbering.

Another embodiment of the present invention provides a method for regulating association in an antibody, further comprising modifying nucleic acids so that amino acid residues of the set of amino acid residues indicated in (d) below are amino acid residues that do not electrically repel each other:

(d) the amino acid residue contained in CH1 at position 213 as indicated by EU numbering, and the amino acid residue contained in CL at position 123 as indicated by EU numbering.

According to the method for regulating association of the present invention, a desired bispecific antibody can be obtained preferentially and efficiently as previously described. Namely, a desired heteromeric multimer in the form of a bispecific antibody can be efficiently formed from a monomer mixture.

The phrase “modify nucleic acids” in the above-mentioned methods of the present invention refers to modifying nucleic acids so that they correspond to amino acid residues introduced by the “modifications” of the present invention. More specifically, it refers to modifying the nucleic acids encoding the original (pre-modified) amino acid residues to the nucleic acids encoding the amino acid residues that are to be introduced by the modification. Ordinarily, it means performing gene manipulations or mutation treatment that would result in at least one nucleotide insertion, deletion, or substitution to the original nucleic acid so that codons encoding amino acid residues of interest is formed. More specifically, codons encoding the original amino acid residues are substituted with codons encoding the amino acid residues that are to be introduced by the modification. Such nucleic acid modification can be performed suitably by those skilled in the art using known techniques such as site-specific mutagenesis and PCR mutagenesis.

In addition, the present invention provides nucleic acids that encode an antibody of the present invention. Moreover, vectors carrying the nucleic acids are also included in the present invention.

The nucleic acids of the present invention are ordinarily carried by (inserted into) suitable vectors and then introduced into host cells. These vectors are not particularly limited so long as the inserted nucleic acid is stably maintained. For example, when using E. coli as the host, the cloning vector is preferably a pBluescript vector (Stratagene) and such, but various commercially available vectors may be used. Expression vectors are particularly useful as vectors for producing the polypeptides of the present invention. Expression vectors are not particularly limited so long as they can express polypeptides in test tubes, E. coli, cultured cells, or individual organisms. For example, preferred vectors include pBEST vector (Promega) for expression in test tubes, pET vector (Invitrogen) for E. coli, pME18S-FL3 vector (GenBank Accession No. AB009864) for cultured cells, and pME18S vector (Mol. Cell Biol. 8:466-472 (1998)) for individual organisms. Insertion of a DNA of the present invention into vectors can be performed using, for example, In-Fusion Advantage PCR Cloning Kit (Clontech).

Further, the present invention provides host cells carrying the above described nucleic acids. The host cells are not particularly limited, and various host cells such as E. coli and various animal cells can be used according to the purpose. The host cells may be used, for example, as a production system to produce and express the antibodies or the polypeptides of the present invention. In vitro and in vivo production systems are available for polypeptide production systems. Production systems that use eukaryotic cells or prokaryotic cells are examples of in vitro production systems.

Eukaryotic cells that can be used as a host cell include, for example, animal cells, plant cells, and fungal cells. Animal cells include: mammalian cells, for example, CHO (J. Exp. Med. (1995) 108:945), COS, 3T3, myeloma, BHK (baby hamster kidney), HeLa, C127, HEK293, Bowes melanoma cells, and Vero; amphibian cells such as Xenopus laevis oocytes (Valle, et al. (1981) Nature 291:338-340); and insect cells (e.g., Drosophila S2, Sf9, Sf21, and Tn5). In the expression of the antibodies of the present invention, CHO-DG44, CHO-DX11B, COST cells, and BHK cells can be suitably used. Among animal cells, CHO cells are particularly preferable for large-scale expression.

Vectors can be introduced into a host cell by known methods, for example, by calcium phosphate methods, the DEAE-dextran methods, methods using cationic liposome DOTAP (Boehringer-Mannheim), electroporation methods (Current protocols in Molecular Biology edit. Ausubel et al. (1987) Publish. John Wiley & Sons, Section 9.1-9.9), lipofection, lipofectamine methods (GIBCO-BRL), or microinjection methods. Moreover, gene introduction to polypeptide expression can also be carried out using the Free Style 293 Expression System (Invitrogen).

Plant cells include, for example, Nicotiana tabacum-derived cells known as a protein production system. Calluses can be cultured from these cells to produce the antibodies of the present invention.

Known protein expression systems are those using fungal cells including yeast cells, for example, cells of genus Saccharomyces such as Saccharomyces cerevisiae and Saccharomyces pombe; and cells of filamentous fungi, for example, cells of genus Aspergillus such as Aspergillus niger. These cells can be used as a host to produce the antibodies of the present invention.

Bacterial cells can be used in the production systems using prokaryotic cells. Examples of bacterial cells include Streptococcus, Staphylococcus, E. coli, Steptomyces, Bacillus subtilis as well as the E. coli described above. Such cells can be used to produce the antibodies of the present invention.

When producing an antibody using a host cell of the present invention, the polynucleotide encoding an antibody of the present invention may be expressed by culturing the host cells transformed with the expression vector containing the polynucleotide. The culture can be performed using known methods. For example, when using animal cells as a host, DMEM, MEM, RPMI 1640, or IMDM may be used as the culture medium, and may be used with serum supplements such as FBS or fetal calf serum (FCS). Serum-free cultures are also acceptable. The preferred pH is about 6 to 8 during the course of culturing. Culture is carried out typically at about 30° C. to 40° C. for about 15 to 200 hours. Medium is exchanged, aerated, or agitated, as necessary.

On the other hand, production systems using animals or plants may be used as systems for producing polypeptides in vivo. A polynucleotide of interest is introduced into an animal or plant and the polypeptide is produced in the body of the animal or plant and then collected. The “host” of the present invention includes such animals and plants.

For secreting host cell-expressed polypeptides into the lumen of the endoplasmic reticulum, periplasmic space, or extracellular environment, suitable secretion signals can be incorporated into the polypeptides of interest. These signals may be intrinsic or foreign to the polypeptides of interest.

When the polypeptides of the present invention are secreted into the culture media, the polypeptides produced by the above-mentioned method can be harvested by collecting the media. When the polypeptides of the present invention are produced inside cells, first, the cells are lysed, and then these polypeptides are collected.

Animals to be used for the production system include mammals and insects. Mammals such as goats, pigs, sheep, mice, and cattle may be used (Vicki Glaser, SPECTRUM Biotechnology Applications (1993)). Alternatively, the mammals may be transgenic animals.

For example, a polynucleotide encoding an antibody of the present invention may be prepared as a fusion gene with a gene encoding a polypeptide specifically produced in milk, such as the goat β-casein. Polynucleotide fragments containing the fusion gene are injected into goat embryos, which are then introduced back to female goats. The desired antibody can be obtained from milk produced by the transgenic goats, which are born from the goats that received the embryos, or from their offspring. Appropriate hormones may be administered to the transgenic goats to increase the volume of milk containing the antibody produced by the transgenic goats (Ebert et al., Bio/Technology 12: 699-702 (1994)).

Insects such as silkworms may also be used for producing the antibodies of the present invention. Baculoviruses carrying a polynucleotide encoding an antibody of interest can be used to infect silkworms, and the antibody of interest can be obtained from their body fluids of (Susumu et al., Nature 315: 592-594 (1985)).

Plants used for producing the antibodies of the present invention include, for example, tobacco. When tobacco is used, a polynucleotide encoding an antibody of interest is inserted into a plant expression vector, for example, pMON 530, and then the vector is introduced into a bacterium, such as Agrobacterium tumefaciens. The bacteria are then used to infect tobacco such as Nicotiana tabacum, and the desired antibodies can be recovered from the tobacco leaves (Ma et al., Eur. J. Immunol. 24: 131-138 (1994)).

The resulting antibody may be isolated from the inside or outside (such as the medium and milk) of host cells, and purified as a substantially pure and homogenous antibody. Methods are not limited to any specific method and any standard method for isolating and purifying antibodies may be used. Antibodies may be isolated and purified, by appropriately selecting and combining, for example, ammonium sulfate or ethanol precipitation, acid extraction, chromatographic columns, filtration, ultrafiltration, salting out, solvent precipitation, solvent extraction, distillation, immunoprecipitation, SDS-polyacrylamide gel electrophoresis, isoelectric focusing, dialysis, recrystallization, and others.

Chromatographies include, for example, affinity chromatographies, ion exchange chromatographies such as anion exchange chromatographies and cation exchange chromatographies, phosphocellulose chromatographies, hydrophobic (interaction) chromatographies, gel filtrations, reverse-phase chromatographies, adsorption chromatographies, hydroxylapatite chromatographies, and lectin chromatographies (Strategies for Protein Purification and Characterization: A Laboratory Course Manual. Ed Daniel R. Marshak et al., Cold Spring Harbor Laboratory Press, 1996). These chromatographies can be carried out using liquid phase chromatographies such as HPLC and FPLC. Examples of the affinity chromatography columns include protein A columns and protein G columns. Examples of the proteins A columns include Hyper D, POROS, and Sepharose F. F. (Pharmacia).

An antibody can be modified freely and peptide portions can be deleted from it by treating the antibody with an appropriate protein modifying enzyme before or after antibody purification, as necessary. Such protein modifying enzymes include, for example, trypsins, chymotrypsins, lysyl endopeptidases, protein kinases, and glucosidases.

In another preferred embodiment, the present invention also includes methods for producing the antibodies of the present invention, such methods including the steps of culturing the host cells of the present invention as described above and collecting the antibodies from such cell culture.

Moreover, the present invention relates to pharmaceutical compositions (pharmaceutical agents) comprising an antibody of the present invention and a pharmaceutically acceptable carrier. In the present invention, pharmaceutical compositions ordinarily refer to pharmaceutical agents for treating or preventing, or testing and diagnosing diseases.

The pharmaceutical compositions of the present invention can be formulated by methods known to those skilled in the art. Moreover, the antibodies of the present invention can be formulated in combination with other pharmaceutical substances, as required. For example, they can be used parenterally in the form of an injection of a sterile solution or suspension with water or another pharmaceutically acceptable liquid. For example, they may be formulated as unit doses that meet the requirements for the preparation of pharmaceuticals by appropriately combining with pharmaceutically acceptable carriers or media, specifically with sterile water, physiological saline, a vegetable oil, emulsifier, suspension, detergent, stabilizer, flavoring agent, excipient, vehicle, preservative, binder, or such. In such preparations, the amount of active ingredient is adjusted such that the dose falls within an appropriately pre-determined range.

Sterile compositions for injection can be formulated using vehicles such as distilled water for injection, according to standard protocols for formulation.

Aqueous solutions for injection include, for example, physiological saline and isotonic solutions containing dextrose or other adjuvants (for example, D-sorbitol, D-mannose, D-mannitol, and sodium chloride). Appropriate solubilizers, for example, alcohols (ethanol and such), polyalcohols (propylene glycol, polyethylene glycol, and such), non-ionic detergents (polysorbate 80™, HCO-50, and such), may be used in combination.

Oils include sesame and soybean oils. Benzyl benzoate and/or benzyl alcohol can be used in combination as solubilizers. Buffers (for example, phosphate buffer and sodium acetate buffer), soothing agents (for example, procaine hydrochloride), stabilizers (for example, benzyl alcohol and phenol), and/or antioxidants can also be combined. Prepared injectables are generally filled into appropriate ampules.

The pharmaceutical compositions of the present invention are preferably administered parenterally. For example, the compositions may be injections, transnasal compositions, transpulmonary compositions or transdermal compositions. For example, such compositions can be administered systemically or locally by intravenous injection, intramuscular injection, intraperitoneal injection, subcutaneous injection, or such.

The administration methods can be appropriately selected in consideration of a patient's age and symptoms. The dose of a pharmaceutical composition containing an antibody or a polynucleotide encoding an antibody may be, for example, from 0.0001 to 1000 mg/kg for each administration. Alternatively, the dose may be, for example, from 0.001 to 100,000 mg per patient. However, the doses are not necessarily limited to the ranges described above. The doses and administration methods vary depending on a patient's weight, age, symptoms, and such. Those skilled in the art can select appropriate doses and administration methods in consideration of the factors described above.

Amino acids contained in the amino acid sequences of the present invention may be post-translationally modified (for example, the modification of an N-terminal glutamine into a pyroglutamic acid by pyroglutamylation is well-known to those skilled in the art). Naturally, such post-translationally modified amino acids are included in the amino acid sequences in the present invention.

All prior art documents cited in the present specification are incorporated herein by reference.

EXAMPLES

Hereinbelow, the present invention will be specifically described with reference to the Examples, but the present invention is not limited thereto.

[Example 1] Search for Sites that Regulate the CH1/CL Interface

It was thought that by introducing mutations into each of the CH1 and CL domains of a bispecific antibody, and utilizing the electrical charge at the CH1/CL interface to regulate the CH1/CL interface, only the H chain and L chain for an antigen A specifically associate, and only the H chain and L chain for an antigen B specifically associate. Hereinafter, it is referred to as regulation of the CH1/CL interface. A search to found out the positions where the CH1/CL interface can be controlled was carried out using a crystal structure model. Amino acids maintain interactions between side chains through hydrophobic interaction, electrical interaction, hydrogen bonding and the like. These interactions are known to occur between side chains present within a range of about 4 Å. Therefore, amino acids were found in a PDB model 1HZH, wherein the distance between amino acids present in CH1 and amino acids present in CL at the interface between CH1 and CL is about 4 Å. The sites at which the amino acids are found are each summarized in FIG. 1 and Table 1 (Summary of Modified Sites). The amino acid numbers shown in Table 1 are indicated in accordance with EU numbering (Sequences of proteins of immunological interest, NIH Publication No. 91-3242). In addition, subsequent amino acid numbers are also indicated in accordance with EU numbering. In the present example, IgG1 was used for the H chain, and IgK (Kappa) was used for the L chain.

TABLE 1 CH CL 1 K147 T180 2 Q175 Q160 3 K213 E123 4 K133 N138 5 K147 S131 6 H168 T164 7 F170 L135

In order to regulate the CH1/CL interface using the electrical charge of amino acids, the amino acids found in CH1 of the H chain or CL of the L chain (Table 1) were substituted with positively charged Lys or His and negatively charged Glu or Asp. More specifically, constant regions: TH2 and TH11, in which amino acids of CH1 of human Gld (SEQ ID NO: 1) were substituted with positively charged Lys; and TH1, TH3, TH4, TH9, TH10, and TH12, in which amino acids of CH1 of human G1d (SEQ ID NO: 1) were substituted with negatively charged Glu or Asp were prepared. Similarly, constant regions: TL2, TL4, TL5, TL6, TL8, and TL12, in which amino acids of human CL (SEQ ID NO: 13) were substituted with positively charged Lys; TL11, in which amino acids of human CL (SEQ ID NO: 13) were substituted with His; and TL1, TL3, TL7, TL9, TL10, and TL13, in which amino acids of human CL (SEQ ID NO: 13) were substituted with negatively charged Glu or Asp were prepared. The names (name), sites of mutation (mutation) and sequence numbers of the prepared constant regions are summarized in Table 2 (Summary of Modified Sites).

TABLE 2 CH1 muta- CL name tion SEQ ID NO name mutation SEQ ID NO TH1 K147E SEQ ID NO: 002 TL1 T180E SEQ ID NO: 014 TH2 Q175K SEQ ID NO: 003 TL2 T180K SEQ ID NO: 015 TH3 Q175E SEQ ID NO: 004 TL3 Q160E SEQ ID NO: 016 TH4 K213E SEQ ID NO: 005 TL4 Q160K SEQ ID NO: 017 TH9 K133E SEQ ID NO: 006 TL5 E123K SEQ ID NO: 018 TH10 H168D SEQ ID NO: 007 TL6 N138K SEQ ID NO: 019 TH11 F170K SEQ ID NO: 008 TL7 N138E SEQ ID NO: 020 TH12 F170E SEQ ID NO: 009 TL8 S131K SEQ ID NO: 021 TL9 S131E SEQ ID NO: 022 TL10 T164D SEQ ID NO: 023 TL11 T164H SEQ ID NO: 024 TL12 L135K SEQ ID NO: 025 TL13 L135E SEQ ID NO: 026

[Example 2] Method for Screening Sites that Regulate the CH1/CL Interface, and Preparation and Analysis of Each Antibody

Effects on the regulation of the CH1/CL interface of the found amino acids were confirmed using the method described below. Screening was carried out using an anti-GPC3 antibody. First, expression vectors of the H chain and L chain were constructed. An H chain expression vector having the H chain variable region GpH7 (SEQ ID NO: 34) and the H chain constant region prepared in Example 1, and an L chain expression vector having the L chain variable region GpL16 (SEQ ID NO: 35) and the L chain constant region prepared in Example 1 were each constructed in accordance with Reference Example 1. Next, combinations of the prepared H chain and L chain expression vectors were selected in the manner described below. A single H chain in which the amino acid at the found site has a positive charge or negative charge was selected from among the constant regions prepared in Example 1. In this case, a mutation was not always introduced. For example, although position 147 of TH1 is substituted with Glu, a mutation is not introduced because the amino acid at position 147 of G1d is Lys and it initially has a positive charge. Next, L chains, which have mutations at the positions corresponding to the mutated positions in CH1 of the selected H chains according Table 1, were selected from Table 2. For example, when TH1 is selected for the H chain, TL1 and TL2 were selected for the L chain, since the amino acid at position 147 of CH1 and the amino acid at position 180 of CL are expected to interact as the CH1/CL interface. Subsequently, the selected two L chains were mixed with the selected one H chain, and antibodies were expressed in accordance with Reference Example 1. Finally, the expressed antibodies were analyzed in accordance with Reference Example 3 or Reference Example 4, and modifications effective for regulating the CH1/CL interface were screened according to the expression ratio of each antibody. Since IgG is composed of a complex of two H chains and two L chains, when one type of H chain and two types of L chains are mixed and expressed, three combinations are expected to be expressed. For example, when combinations of TH1 as the H chain, and TL1 and TL2 as the L chains are expressed, three combinations below are expressed (FIG. 2): H chain_1:H chain_2: L chain_1:L chain_2=TH1:TH1:TL1:TL1 (indicated as TH1/TL1), H chain_1:H chain_2: L chain_1:L chain_2=TH1:TH1:TL1:TL2 (indicated as TH1/TL1_TL2) H chain_1:H chain_2: L chain_1:L chain_2=TH1:TH1:TL2:TL2 (indicated as TH1/TL2). In the case that association of the H chain and L chain is not selective, the H chain and L chains are expected to be expressed in the ratio TH1/TL1:TH1/TL1_TL2:TH1/TL2=1:2:1, since the two L chains are present in equal amounts. However, in the case that the H chain preferentially binds to only either one of the L chains at the CH1/CL interface, it is thought that only that combination is expressed preferentially. For example, in the case that the amino acid at position 147 of CH1 and the amino acid at position 180 of CL are involved at the CH1/CL interface, when TH1 is expressed as the H chain, and TL1 and TL2 are expressed as the L chains, the combination of TH1, in which the amino acid at position 147 of CH1 is Glu (negatively charged), and TL2, in which the amino acid at position 180 of the L chain CL is Lys (positively charged), is expected to be expressed preferentially. However, in the case that the amino acid at position 147 of CH1 and the amino acid at position 180 of CL are not interacting on the CH1/CL interface, since the association of the H chain and L chain is not selective, they are expected to be expressed in the ratio of TH1/TL1:TH1/TL1_TL2:TH1/TL2=1:2:1. In this manner, modifications effective for regulation of the CH1/CL interface (modifications involved in the CH1/CL interface) were screened by mixing and expressing one type of H chain and two types of L chains, and using the expression balance thereof as an indicator.

Combinations of H chain and L chains are summarized in Table 3 (Combinations of H Chain and L Chains Used in Expression; Sites of Mutation in H Chain and L Chain also Shown).

TABLE 3 Hch Lch name mutation SEQ ID NO name mutation SEQ ID NO G1d K147K SEQ ID NO: 001 TL1_TL2 T180E_T180K SEQ ID NO: 014, SEQ ID NO: 015 TH1 K147E SEQ ID NO: 002 TL1_TL2 T180E_T180K SEQ ID NO: 014, SEQ ID NO: 015 TH2 Q175K SEQ ID NO: 003 TL3_TL4 Q160E_Q160K SEQ ID NO: 016, SEQ ID NO: 017 TH3 Q175E SEQ ID NO: 004 TL3_TL4 Q160E_Q160K SEQ ID NO: 016, SEQ ID NO: 017 G1d K213K SEQ ID NO: 001 k0_TL5 E123E_E123K SEQ ID NO: 013, SEQ ID NO: 018 TH4 K213E SEQ ID NO: 005 k0_TL5 E123E_E123K SEQ ID NO: 013, SEQ ID NO: 018 G1d K133K SEQ ID NO: 001 TL6_TL7 N138K_N138E SEQ ID NO: 019, SEQ ID NO: 020 TH9 K133E SEQ ID NO: 006 TL6_TL7 N138K_N138E SEQ ID NO: 019, SEQ ID NO: 020 G1d K147K SEQ ID NO: 001 TL8_TL9 S131K_S138E SEQ ID NO: 021, SEQ ID NO: 022 TH1 K147E SEQ ID NO: 002 TL8_TL9 S131K_S138E SEQ ID NO: 021, SEQ ID NO: 022 G1d H168H SEQ ID NO: 001 TL10_TL11 T164D_T164H SEQ ID NO: 023, SEQ ID NO: 024 TH10 H168D SEQ ID NO: 007 TL10_TL11 T164D_T164H SEQ ID NO: 023, SEQ ID NO: 024 TH11 F170K SEQ ID NO: 008 TL12_TL13 L135K_L135E SEQ ID NO: 025, SEQ ID NO: 026 TH12 F170E SEQ ID NO: 009 TL12_TL13 L135K_L135E SEQ ID NO: 025, SEQ ID NO: 026

Antibodies were expressed in accordance with the combinations shown in Table 3, and the effects on regulation of the selected CH1/CL interface were confirmed. At that time, antibodies of one type of H chain and one type of L chain were simultaneously expressed, and used as a control in analyses. Antibodies were expressed in accordance with the method of Reference Example 1.

The prepared antibodies were analyzed by AIEX in accordance with the method of Reference Example 3. Data of the AIEX analysis are summarized in FIG. 3. Since an anion exchange column is used in the AIEX analysis, positively charged antibodies are eluted more rapidly. The analyzed data are summarized and shown in Table 4. The peaks are indicated as peaks 1, 2, and 3 in the order of increasing elution time. The ratio of each peak was calculated with the total of the peak areas being 100%.

As shown in FIG. 2, in the case that the positions introduced with mutations form electrical interaction, the ratio of antibody at the position of the gray-colored peak increases. Namely, mutation sites at which the ratio of the gray-colored antibody is greater than 25% are thought to be interacting electrically.

TABLE 4 Hch Lch peak1 peak2 peak3 name mutation name mutation % % % G1d K147K TL1 T180E 100 G1d K147K TL2 T180K 100 G1d K147K TL1_TL2 T180E_T180K 20.2 44.2 35.6 TH1 K147E TL1 T180E 100 TH1 K147E TL2 T180K 100 TH1 K147E TL1_TL2 T180E_T180K 28.5 46.7 24.8 TH2 Q175K TL3 Q160E 100 TH2 Q175K TL4 Q160K 100 TH2 Q175K TL3_TL4 Q160E_Q160K 13 44 43 TH3 Q175E TL3 Q160E 100 TH3 Q175E TL4 Q160K 100 TH3 Q175E TL3_TL4 Q160E_Q160K 25 49.9 24.8 G1d K213K k0 E123E 100 G1d K213K TL5 E123K 100 G1d K213K k0_TL5 E123E_E123K 15.1 39.3 45.6 TH4 K213E k0 E123E 100 TH4 K213E TL5 E123K 100 TH4 K213E k0_TL5 E123E_E123K 17.5 41.3 41.2 G1d K133K TL6 N138K 100 G1d K133K TL7 N138E 100 G1d K133K TL6_TL7 N138K_N138E 27.3 44 28.7 TH9 K133E TL6 N138K 100 TH9 K133E TL7 N138E 100 TH9 K133E TL6_TL7 N138K_N138E 29.1 44 26.9 G1d K213K TL8 S131K 100 G1d K213K TL9 S131E 100 G1d K213K TL8_TL9 S131K_S131E 17.8 45.7 36.7 TH1 K147E TL8 S131K 100 TH1 K147E TL9 S131E 100 TH1 K147E TL8_TL9 S131K_S131E 36.9 44.1 19 G1d H168H TL10 T164D 100 G1d H168H TL11 T164H 100 G1d H168H TL10_TL11 T164D_T164H 27 43.3 29.7 TH10 H168D TL10 T164D 100 TH10 H168D TL11 T164H 100 TH10 H168D TL10_TL11 T164D_T164H 27 44.4 28.6 TH11 F170K TL12 L135K 100 TH11 F170K TL13 L135E 100 TH11 F170K TL12_TL13 L135K_L135E not expressed TH12 F170E TL12 L135K 100 TH12 F170E TL13 L135E 100 TH12 F170E TL12_TL13 L135K_L135E 38.1 41.6 20.3

As a result of examining various sites of modification in this manner, positions 147, 175, and 213 of the H chain, and positions 123, 131, 160, and 180 of the L chain were thought to be effective for regulating the CH1/CL interface. In addition, it was found that modifications of only position 147 of the H chain and position 123 of the L chain reported in WO 2006/106905 and WO 2007/147901 were inadequate for causing specific association of the H chain and L chain, and the specific association is possible only by combining modifications found in the present example.

[Example 3] Preparation and Analysis of Antibodies with Combined Sites of Modification

It was thought that the CH1/CL interface is regulated more effectively by combining the sites of K147, Q175, and K213 in CH1 and the sites of E123, 5131, Q160, and T180 in CL as found in Example 2, which were thought to have considerable effects in regulating the CH1/CL interface. The combinations of modifications in the prepared antibodies, and the expressed antibodies are summarized in Table 5 (Summary of Modification Sites).

TABLE 5 Hch Lch name mutation SEQ ID NO name mutation SEQ ID NO G1d K147K SEQ ID NO: 001 TL14_TL15 T180K, S131K_T180E, S131E SEQ ID NO: 027, SEQ ID NO: 028 TH1 K147E SEQ ID NO: 002 TL14_TL15 T180K, S131K_T180E, S131E SEQ ID NO: 027, SEQ ID NO: 028 TH2 Q175K SEQ ID NO: 003 TL16_TL17 T180K, S131K, Q160K_T180E, S131E, Q160E SEQ ID NO: 029, SEQ ID NO: 030 TH13 K147E, Q175E SEQ ID NO: 010 TL16_TL17 T180K, S131K, Q160K_T180E, S131E, Q160E SEQ ID NO: 029, SEQ ID NO: 030 G1d K147K SEQ ID NO: 001 TL18_TL15 T180K, S131K, E123K_T180E, S131E, E123E SEQ ID NO: 031, SEQ ID NO: 028 TH14 K147E, K213E SEQ ID NO: 011 TL18_TL15 T180K, S131K, E123K_T180E, S131E, E123E SEQ ID NO: 031, SEQ ID NO: 028 TH2 Q175K SEQ ID NO: 003 TL19_TL17 T180K, S131K, Q160K, E123K_T180E, S131E, SEQ ID NO: 032, SEQ ID NO: 030 Q160E TH15 K147E, Q175E, SEQ ID NO: 012 TL19_TL17 T180K, S131K, Q160K, E123K_T180E, S131E, SEQ ID NO: 032, SEQ ID NO: 030 K213E Q160E, E123E

Preparation of expression vectors of the H chain or L chain introduced with mutations, and antibody expression were carried out in accordance with Reference Example 1, and analyses of the prepared antibodies were carried out in accordance with Reference Example 3 or Reference Example 4. The results are summarized in Table 6.

TABLE 6 Hch Lch peak 1 peak 2 peak 3 name mutation name mutation % % % G1d K147K TL14 T180K, S131K 100 G1d K147K TL15 T180E, S131E 100.0 G1d K147K TL14_TL15 T180K, S131K, T180E, S131E 11.5 43.6 44.9 TH1 K147E TL14 T180K, S131K 100.0 TH1 K147E TL15 T180E, S131E 100.0 TH1 K147E TL14_TL15 T180K, S131K_T180E, S131E 46.1 43.4 10.4 TH2 Q175K TL16 T180K, S131K, Q160K not expressed TH2 Q175K TL17 T180E, S131E, Q160E 100.0 TH2 Q175K TL16_TL17 T180K, S131K, Q160K_T180E, 1.4 16.5 82.0 S131E, Q160E TH13 K147E, Q175E TL16 T180K, S131K, Q160K 100.0 TH13 K147E, Q175E TL17 T180E, S131E, Q160E not expressed TH13 K147E, Q175E TL16_TL17 T180K, S131K, Q160K_T180E, 70.2 26.4 3.4 S131E, Q160E G1d K147K TL18 T180K, S131K, E123K not expressed G1d K147K TL15 T180E, S131E 100.0 G1d K147K TL18_TL15 T180K, S131K, E123K_T180E, 5.1 35.3 59.6 S131E, E123E TH14 K147E, K213E TL18 T180K, S13SK, E123K 100.0 TH14 K147E, K213E TL15 T180E, S131E 100.0 TH14 K147E, K213E TL18_TL15 T180K, S131K, E123K_T180E, 44.5 44.4 11.1 S131E, E123E TH2 Q175K TL19 T180K, S131K, Q160K, E123K not expressed TH2 Q175K TL17 T180E, S131E, Q160E 100.0 TH2 Q175K TL19_TL17 T180K, S131K, Q160K, 93.1 E123K_T180E, S131E, Q160E TH15 K147E, Q175E, TL19 T180K, S131K, Q160K, E123K 100.0 K213E TH15 K147E, Q175E, TL17 T180E, S131E, Q160E not K213E expressed TH15 K147E, Q175E, TL19_TL17 T180K, S131K, Q160K, 78.1 19.9 2.0 K213E E123K_T180E, S131E, Q160E

When Table 4 and Table 6 are compared, it is understood that the ratio of targeted combinations of the H chain and L chain is increased by combining modifications, as compared with the introduction of a single modification. Consequently, it is thought that an antibody in which only the H chain and L chain of interest have associated can be efficiently prepared by combining modifications.

[Example 4] Expression and Analysis of Bispecific Antibodies

In Example 3, preparation of bispecific antibodies (bispecific Abs) for TH2, TH13, and TH15 of H chains, and TL16, TL17, TL19, and TL20 of L chains that showed considerable effects for regulating the CH1/CL interface was taken thought. In this example, bispecific antibodies were prepared using an anti-IL6R antibody and an anti-GPC3 antibody.

The constant regions of H chain (SEQ ID NO: 59) and L chain (SEQ ID NO: 60) recognizing anti-IL6R, and the constant regions of H chain (SEQ ID NO: 61) and L chain (SEQ ID NO: 62) recognizing anti-GPC3 were substituted with TH2, TH13, and TH15 for the constant regions of the H chain, and with TL16, TL17, TL19, and TL20 for CL of the L chain. Moreover, an H chain introduced with a Knob into Hole (KiH) modification (WO 96/27011) was prepared to avoid association between homogeneous H chains. The mutation sites in these prepared antibodies and the expressed antibodies are summarized in Table 7 (Combinations of H Chain and L Chain of Each Bispecific Antibody).

TABLE 7 Ach Bch NAME VH CH VL CL VH CH VL CL comment 4ch_001 MH0 G1d ML0 k0 GpH7 G1d GpL16 k0 4ch_002 MH0 G1dk ML0 k0 GpH7 G1dh GpL16 k0 KiH 4ch_003 MH0 TH2 ML0 TL17 GpH7 TH13 GpL16 TL16 CH1/CL_1 4ch_004 MH0 TH2k ML0 TL17 GpH7 TH13h GpL16 TL16 KiH + CH1/CL_1 4ch_011 MH0 TH2k ML0 TL17 GpH7 TH13h ML0 TL17 4ch_012 MH0 TH2k GpL 16 TL16 GpH7 TH13h GpL16 TL16 4ch_005 MH0 TH2 ML0 TL17 GpH7 TH15 GpL16 TL19 CH1/CL_2 4ch_006 MH0 TH2k ML0 TL17 GpH7 TH15h GpL16 TL19 KiH + CH1/CL_2 4ch_015 MH0 TH2k ML0 TL17 GpH7 TH15h ML0 TL17 4ch_016 MH0 TH2k GpL 16 TL19 GpH7 TH15h GpL16 TL19 4ch_001 MH0 G1d ML0 k0 GdH7 G1d GpL16 k0 4ch_002 MH0 G1dk ML0 k0 GpH7 G1dh GpL16 k0 KiH 4ch_007 MH0 TH13 ML0 TL16 GpH7 TH2 GpL16 TL17 CH1/CL_1 4ch_008 MH0 TH13k ML0 TL16 GpH7 TH2h GpL16 TL17 KiH + CH1/CL_1 4ch_013 MH0 TH13k ML0 TL16 GpH7 TH2h ML0 TL16 4ch_014 MH0 TH13k GpL 16 TL17 GpH7 TH2h GpL16 TL17 4ch_009 MH0 TH15 ML0 TL19 GpH7 TH2 GpL16 TL17 CH1/CL_2 4ch_010 MH0 TH15k ML0 TL19 GpH7 TH2h GpL16 TL17 KiH + CH1/CL_2 4ch_017 MH0 TH15k ML0 TL19 GpH7 TH2h ML0 TL19 4ch_018 MH0 TH15k GpL 16 TL17 GpH7 TH2h GpL16 TL17

In Table 7 above, “k” is added after the variant name for those constant regions in which a “knob” modification was introduced into the H chain, and “h” is added after the variant name for those constant regions in which a “hole” modification was introduced. For example, “THlk” indicates that a “knob” modification was introduced in addition to the TH1 mutation, and “TH1h” indicates that a “hole” modification was introduced in addition to the TH1 mutation. Preparation of expression vectors of the H chain or L chain into which a mutation has been introduced as well as expression of antibodies were carried out in accordance with Reference Example 1, and analyses of the prepared antibodies were carried out in accordance with the CIEX analysis method shown in Reference Example 4.

The combinations of H chains and L chains of the anti-IL6R antibody and anti-GPC3 antibody used in the bispecific antibodies are summarized in Table 8.

TABLE 8 VH CH VL CL NAME chain name mutation SEQ ID NO name mutation SEQ ID NO name mutation SEQ ID NO name mutation SEQ ID NO 4ch_001 Ach MH0 SEQ ID NO: 036 G1d K147K SEQ ID NO: 001 ML0 SEQ ID NO: 037 k0 SEQ ID NO: 013 4ch_002 Ach MH0 SEQ ID NO: 036 G1dk SEQ ID NO: 038 ML0 SEQ ID NO: 037 k0 SEQ ID NO: 013 4ch_003 Ach MH0 SEQ ID NO: 036 TH2 Q175K SEQ ID NO: 003 ML0 SEQ ID NO: 037 TL17 T180E, S131E, Q160E SEQ ID NO: 030 4ch_004 Ach MH0 SEQ ID NO: 036 TH2k Q175K SEQ ID NO: 039 ML0 SEQ ID NO: 037 TL17 T180E, S131E, Q160E SEQ ID NO: 030 4ch_011 Ach MH0 SEQ ID NO: 036 TH2k Q175K SEQ ID NO: 039 ML0 SEQ ID NO: 037 TL17 T180E, S131E, Q160E SEQ ID NO: 030 4ch_012 Ach MH0 SEQ ID NO: 036 TH2k Q175K SEQ ID NO: 039 GpL16 SEQ ID NO: 035 TL16 T180K, S131K, Q160K SEQ ID NO: 029 4ch_005 Ach MH0 SEQ ID NO: 036 TH2 Q175K SEQ ID NO: 003 ML0 SEQ ID NO: 037 TL17 T180E, S131E, Q160E SEQ ID NO: 030 4ch_006 Ach MH0 SEQ ID NO: 036 TH2k Q175K SEQ ID NO: 039 ML0 SEQ ID NO: 037 TL17 T180E, S131E, Q160E SEQ ID NO: 030 4ch_015 Ach MH0 SEQ ID NO: 036 TH2k Q175K SEQ ID NO: 039 ML0 SEQ ID NO: 037 TL17 T180E, S131E, Q160E SEQ ID NO: 030 4ch_016 Ach MH0 SEQ ID NO: 036 TH2k Q175K SEQ ID NO: 039 GpL16 SEQ ID NO: 035 TL19 T180K, S131K, Q160K, SEQ ID NO: 032 E123K 4ch_001 Ach MH0 SEQ ID NO: 036 G1d K147K SEQ ID NO: 001 ML0 SEQ ID NO: 037 k0 SEQ ID NO: 013 4ch_002 Ach MH0 SEQ ID NO: 036 G1dk SEQ ID NO: 038 ML0 SEQ ID NO: 037 k0 SEQ ID NO: 013 4ch_007 Ach MH0 SEQ ID NO: 036 TH13 K147E, Q175E SEQ ID NO: 010 ML0 SEQ ID NO: 037 TL16 T180K, S131K, Q160K SEQ ID NO: 029 4ch_008 Ach MH0 SEQ ID NO: 036 TH13k K147E, Q175E SEQ ID NO: 040 ML0 SEQ ID NO: 037 TL16 T180K, S131K, Q160K SEQ ID NO: 029 4ch_013 Ach MH0 SEQ ID NO: 036 TH13k K147E, Q175E SEQ ID NO: 040 ML0 SEQ ID NO: 037 TL16 T180K, S131K, Q160K SEQ ID NO: 029 4ch_014 Ach MH0 SEQ ID NO: 036 TH13k K147E, Q175E SEQ ID NO: 040 GpL16 SEQ ID NO: 035 TL17 T180E, S131E, Q160E SEQ ID NO: 030 4ch_009 Ach MH0 SEQ ID NO: 036 TH15 K147E, Q175E, K213E SEQ ID NO: 012 ML0 SEQ ID NO: 037 TL19 T180K, S131K, Q160K, SEQ ID NO: 032 E123K 4ch_010 Ach MH0 SEQ ID NO: 036 TH15k K147E, Q175E, K213E SEQ ID NO: 041 ML0 SEQ ID NO: 037 TL19 T180K, S131K, Q160K, SEQ ID NO: 032 E123K 4ch_017 Ach MH0 SEQ ID NO: 036 TH15k K147E, Q175E, K213E SEQ ID NO: 041 ML0 SEQ ID NO: 037 TL19 T180K, S131K, Q160K, SEQ ID NO: 032 E123K 4ch_018 Ach MH0 SEQ ID NO: 036 TH15k K147E, Q175E, K213E SEQ ID NO: 041 GpL16 SEQ ID NO: 035 TL17 T180E, S131E, Q160E SEQ ID NO: 030 4ch_001 Bch GpH7 SEQ ID NO: 034 G1d K147K SEQ ID NO: 001 GpL16 SEQ ID NO: 035 k0 SEQ ID NO: 013 4ch_002 Bch GpH7 SEQ ID NO: 034 G1dh SEQ ID NO: 042 GpL16 SEQ ID NO: 035 k0 SEQ ID NO: 013 4ch_003 Bch GpH7 SEQ ID NO: 034 TH13 K147E, Q175E SEQ ID NO: 010 GpL16 SEQ ID NO: 035 TL16 T180K, S131K, Q160K SEQ ID NO: 029 4ch_004 Bch GpH7 SEQ ID NO: 034 TH13h K147E, Q175E SEQ ID NO: 044 GpL16 SEQ ID NO: 035 TL16 T180K, S131K, Q160K SEQ ID NO: 029 4ch_011 Bch GpH7 SEQ ID NO: 034 TH13h K147E, Q175E SEQ ID NO: 044 ML0 SEQ ID NO: 037 TL17 T180E, S131E, Q160E SEQ ID NO: 030 4ch_012 Bch GpH7 SEQ ID NO: 034 TH13h K147E, Q175E SEQ ID NO: 044 GpL16 SEQ ID NO: 035 TL16 T180K, S131K, Q160K SEQ ID NO: 029 4ch_005 Bch GpH7 SEQ ID NO: 034 TH15 K147E, Q175E, K213E SEQ ID NO: 012 GpL16 SEQ ID NO: 035 TL19 T180K, S131K, Q160K, SEQ ID NO: 032 E123K 4ch_006 Bch GpH7 SEQ ID NO: 034 TH15h K147E, Q175E, K213E SEQ ID NO: 045 GpL16 SEQ ID NO: 035 TL19 T180K, S131K, Q160K, SEQ ID NO: 032 E123K 4ch_015 Bch GpH7 SEQ ID NO: 034 TH15h K147E, Q175E, K213E SEQ ID NO: 045 ML0 SEQ ID NO: 037 TL17 T180E, S131E, Q160E SEQ ID NO: 030 4ch_016 Bch GpH7 SEQ ID NO: 034 TH15h K147E, Q175E, K213E SEQ ID NO: 045 GpL16 SEQ ID NO: 035 TL19 T180K, S131K, Q160K, SEQ ID NO: 032 E123K 4ch_001 Bch GpH7 SEQ ID NO: 034 G1d K147K SEQ ID NO: 001 GpL16 SEQ ID NO: 035 k0 SEQ ID NO: 013 4ch_002 Bch GpH7 SEQ ID NO: 034 G1dh SEQ ID NO: 042 GpL16 SEQ ID NO: 035 k0 SEQ ID NO: 013 4ch_007 Bch GpH7 SEQ ID NO: 034 TH2 Q175K SEQ ID NO: 003 GpL16 SEQ ID NO: 035 TL17 T180E, S131E, Q160E SEQ ID NO: 030 4ch_008 Bch GpH7 SEQ ID NO: 034 TH2h Q175K SEQ ID NO: 043 GpL16 SEQ ID NO: 035 TL17 T180E, S131E, Q160E SEQ ID NO: 030 4ch_013 Bch GpH7 SEQ ID NO: 034 TH2h Q175K SEQ ID NO: 043 ML0 SEQ ID NO: 037 TL16 T180K, S131K, Q160K SEQ ID NO: 029 4ch_014 Bch GpH7 SEQ ID NO: 034 TH2h Q175K SEQ ID NO: 043 GpL16 SEQ ID NO: 035 TL17 T180E, S131E, Q160E SEQ ID NO: 030 4ch_009 Bch GpH7 SEQ ID NO: 034 TH2 Q175K SEQ ID NO: 003 GpL16 SEQ ID NO: 035 TL17 T180E, S131E, Q160E SEQ ID NO: 030 4ch_010 Bch GpH7 SEQ ID NO: 034 TH2h Q175K SEQ ID NO: 043 GpL16 SEQ ID NO: 035 TL17 T180E, S131E, Q160E SEQ ID NO: 030 4ch_017 Bch GpH7 SEQ ID NO: 034 TH2h Q175K SEQ ID NO: 043 ML0 SEQ ID NO: 037 TL19 T180K, S131K, Q160K, SEQ ID NO: 032 E123K 4ch_018 Bch GpH7 SEQ ID NO: 034 TH2h Q175K SEQ ID NO: 043 GpL16 SEQ ID NO: 035 TL17 T180E, S131E, Q160E SEQ ID NO: 030

Each combination is explained herein using 4ch_001, 4 ch_002, 4 ch_003, 4ch_004, 4 ch_011, and 4ch_012 as examples. Modifications that regulate the CH1/CL interface are summarized in Table 8.

4ch_001 was expressed using H chains and L chains that do not have the introduction of modifications for regulating the CH1/CL interface and the KiH modification. 4ch_002 was expressed using H chains and L chain into which the KiH modification was introduced. 4ch_003 was expressed using H chains and L chains into which modifications for regulating the CH1/CL interface were introduced. 4ch_004 was expressed using H chains and an L chains into which the KiH modification and modifications for regulating the CH1/CL interface were introduced. In addition, 4ch_011 was expressed using the H chains of an anti-IL6R antibody and the H chains of an anti-GPC3 antibody into which the KiH modification and modifications for regulating the CH1/CL interface were introduced, and the L chains of an anti-IL6R antibody into which modifications for regulating the CH1/CL interface were introduced. 4ch_012 was expressed using the H chains of an anti-IL6R antibody and the H chains of an anti-GPC3 antibody into which the KiH modification and modifications for regulating the CH1/CL interface were introduced, and the L chains of an anti-GPC3 antibody into which modifications for regulating the CH1/CL interface were introduced. Each antibody was expressed in accordance with Reference Example 1, and analyzed by CIEX in accordance with Reference Example 4; and the results are summarized in FIGS. 9-1 and 9-2. The case of using the H-chain variable region of an anti-IL6R antibody is indicated with MHO, and the case of using the H chain variable region of an anti-GPC3 antibody is indicated with GpH7. The case of using the L chain variable region of an anti-IL6R antibody is indicated with ML0, and the case of using the L chain variable region of an anti-GPC3 antibody is indicated with GpL16. Multiple heterogeneous components which are thought to be various combinations of H chain and L chain were detected by chromatography for 4ch_001 which is not introduced with the mutations for CH1/CL interface control and KiH. In contrast, since the association of homogeneous H chains was suppressed in 4ch_002 with KiH mutation, the number of chromatographic peaks which are thought to be impurities are reduced. In addition, since the association of H chains and L chains was suppressed in 4ch_003 which uses TH2 and TH13 in H chains and TL16 and TL17 in L chains, into which modifications for regulating the CH1/CL interface were introduced, the number of chromatography peaks which are thought to represent impurities decreased. Moreover, it was revealed that 4ch_004 combining the mutations of KiH and modifications for regulating the CH1/CL interface is mostly the main peak. The reason that the chromatography peaks of 4ch_004 nearly coincide with the chromatography peaks of 4ch_011 is thought to be that their peaks are unable to be separated by chromatography due to their similar isoelectric points (pI). Studies were also conducted on 4ch_005, 4ch_006, 4ch_015, and 4ch_016, to which a different regulation of the CH1/CL interface from that for 4ch_004 had been applied; 4ch_007, 4ch_008, 4ch_013, and 4ch_014, in which modifications for regulating the CH1/CL interface introduced into 4ch_004 were interchanged between the anti-GPC3 antibody and the anti-IL6R antibody; and 4ch_009, 4ch_010, 4ch_017, and 4ch_018, in which modifications for regulating the CH1/CL interface introduced into 4ch_006 were interchanged between the anti-GPC3 antibody and the anti-IL6R antibody, using the same methods as 4ch_003, 4ch_004, 4ch_011, and 4ch_012. As a result, heterogeneous components presented in chromatogram were reduced significantly in comparison with 4ch_001.

From the above, it became apparent that bispecific antibodies can be efficiently prepared by combining modifications for regulating the CH1/CL interface and the KiH modification.

[Example 5] Effects of Modifying Regulation of the CH1/CL Interface Using Different Antibodies

It became apparent from Example 4 that the interface regulation using CH1/CL is useful for preparing bispecific antibodies. Therefore, the effect of regulating the CH1/CL interface was confirmed using an anti-CD3 antibody, M12 (H chain: SEQ ID NO: 54, L chain: SEQ ID NO: 57) and an anti-GPC3 antibody, GC33(2) (H chain: SEQ ID NO: 55, L chain: SEQ ID NO: 58). As with Example 4, bispecific antibodies were prepared using TH2, TH13, and TH15 in H chains and TL16, TL17, and TL19 in L chains which demonstrated considerable effects for regulating the CH1/CL interface.

The constant regions of the H chain (SEQ ID NO: 54) and the L chain (SEQ ID NO: 57) of anti-CD3 recognizing antibody M12, and the constant regions of the H chain (SEQ ID NO: 55) and the L chain (SEQ ID NO: 58) of anti-GPC3 recognizing antibody GC33(2), were substituted with TH2, TH13, and TH15 for CH1 of the H chain, and with TL16, TL17, and TL19 for CL of the L chain. Moreover, an H chain with Knob into Hole (KiH) modifications (Patent Document 1) was prepared to avoid association between homogeneous H chains. The mutation sites of these prepared antibodies and the expressed antibodies are summarized in Table 9 (Summary of Modification Sites).

Ach Bch NAME VH CH VL CL VH CH VL CL 4ch_001 M12VH G1d M12VL k0 G1d k0 4ch_004 M12VH TH2k M12VL TL17 TH h TL16 4ch_ M12VH TH2k M12VL TL17 TH h TL19 4ch_001 MH0 G1d ML0 k0 GpH7 G1d GpL16 k0 4ch_ MH0 TH13k ML0 TL18 GpH7 TH2h GpL16 TL17 4ch_ MH0 TH15k ML0 TL19 GdH7 TH2h GpL16 TL17 VH CH NAME chain name mutation SEQ ID NO name mutation SEQ ID NO 4ch_001 Ach M12VH SEQ ID NO: SEQ ID NO: 4ch_004 Ach M12VH SEQ ID NO: Q175K SEQ ID NO: 4ch_ Ach M12VH SEQ ID NO: Q175K SEQ ID NO: 4ch_001 Ach MH0 SEQ ID NO: SEQ ID NO: 4ch_ Ach MH0 SEQ ID NO: SEQ ID NO: 4ch_010 Ach MH0 SEQ ID NO: SEQ ID NO: 4ch_001 Bch SEQ ID NO: SEQ ID NO: 4ch_004 Bch SEQ ID NO: SEQ ID NO: 4ch_ Bch SEQ ID NO: SEQ ID NO: 4ch_001 Bch GpH7 SEQ ID NO: SEQ ID NO: 4ch_ Bch GpH7 SEQ ID NO: Q175K SEQ ID NO: 4ch_010 Bch GpH7 SEQ ID NO: Q175K SEQ ID NO: VL CL NAME chain name mutation SEQ ID NO name mutation SEQ ID NO 4ch_001 Ach M12VL SEQ ID NO: k0 SEQ ID NO: 4ch_004 Ach M12VL SEQ ID NO: TL17 SEQ ID NO: 4ch_ Ach M12VL SEQ ID NO: TL17 SEQ ID NO: 4ch_001 Ach MH0 SEQ ID NO: k0 SEQ ID NO: 4ch_ Ach ML0 SEQ ID NO: TL18 SEQ ID NO: 4ch_010 Ach ML0 SEQ ID NO: TL19 SEQ ID NO: 4ch_001 Bch SEQ ID NO: k0 SEQ ID NO: 4ch_004 Bch SEQ ID NO: TL1 SEQ ID NO: 4ch_ Bch SEQ ID NO: TL19 SEQ ID NO: 4ch_001 Bch GpL16 SEQ ID NO: k0 SEQ ID NO: 4ch_ Bch GpL16 SEQ ID NO: TL17 SEQ ID NO: 4ch_010 Bch GpL16 SEQ ID NO: TL17 SEQ ID NO: indicates data missing or illegible when filed

Preparation of expression vectors of H chains and L chains with mutations and expression of antibodies were carried out in accordance with Reference Example 1, and analyses of the prepared antibodies were carried out in accordance with the CIEX analysis method shown in Reference Example 4.

It is apparent that regulation of the CH1/CL interface is also useful for preparing bispecific antibodies with an anti-CD3 antibody and an anti-GPC3 antibody.

[Example 5] Combination of Regulation of the CH1/CL Interface and Regulation of the Variable Region Interface

When preparing bispecific antibodies, introducing electrical repulsion into the variable regions VH and VL is known as a technique for allowing specific association of target H chains and L chains (Patent Document WO 2006/106905). Therefore, in order to efficiently express only target components, one is to cause repulsion between variable regions of the H chain and L chain, in addition to regulation of the CH1/CL interface. This is referred to as VH/VL interface regulation. MH01 (SEQ ID NO: 46), in which Gln at position 39 as indicated by Kabat numbering of the H chain of anti-IL6R was substituted with Lys; MH02 (SEQ ID NO: 47), in which the Gln was substituted with Glu; ML01 (SEQ ID NO: 50), in which Gln at position 38 as indicated by Kabat numbering of the L chain was substituted with Glu; and ML02 (SEQ ID NO: 51), in which the Glu was substituted with Lys, were prepared. Moreover, GpH71 (SEQ ID NO: 48), in which Gln at position 39 as indicated by Kabat numbering of the H chain of anti-GPC3 was substituted with Lys; GpH72 (SEQ ID NO: 49), in which the Gln was substituted with Glu; GpL161 (SEQ ID NO: 52), in which Gln at position 38 as indicated by Kabat numbering of the L chain was substituted with Glu; and GpL162 (SEQ ID NO: 53), in which the Gln was substituted with Lys, were respectively prepared. Preparation of antibody expression vectors was carried out in accordance with the method of Reference Example 1. Bispecific antibodies were expressed using the prepared antibodies. The combinations of modifications in the prepared antibodies and the expressed antibodies are summarized in Table 10.

TABLE 10 VH CH VL CL SEQ SEQ SEQ SEQ ID ID ID ID NAME chain name mutation NO name mutation NO name mutation NO name mutation NO 4ch_1,2_004 Ach MH01 Q39K 46 TH2k Q175K 39 MH01 Q38E 50 TL17 T180E, S131E, 30 Q160E 4ch_1,2_006 Ach MH01 O39K 46 TH2k Q175K 39 MH01 Q38E 50 TL17 T180E, S131E, 30 Q160E 4ch_2,1_008 Ach MH02 Q39E 47 TH13k K147E, Q175E 49 MH02 Q38K 51 TL16 T180K, S131K, 29 Q160K 4ch_2,1_010 Ach MH02 Q39E 47 TH15k K147E, Q175E, 41 MH02 Q38K 51 TL19 T180K, S131K, 32 K213E Q160K, E123K 4ch_1,2_004 Bch GpH72 Q39E 49 TH13h K147E, Q175E 44 GpL162 Q38K 53 TL16 T180K, S131K, 29 Q160K 4ch_1,2_006 Bch GpH72 Q39E 49 TH15h K147E, Q175E, 45 GpL162 Q38K 53 TL19 T180K, S131K, 32 K213E Q160K, E123K 4ch_2,1_008 Bch GpH71 Q39K 48 TH2h Q175K 43 GpL161 Q38E 52 TL17 T180E, S131E, 30 Q160E 4ch_2,1_010 Bch GpH71 Q39K 48 TH2h Q175K 43 GpL161 Q38E 52 TL17 T180E, S131E, 30 Q160E

Antibody expression was carried out in accordance with Reference Example 1, and analyses of the prepared antibodies were carried out in accordance with Reference Example 4.

Since the peaks observed in the chromatograms of 4ch_006 and 4ch_008, into which mutations for regulating the CH1/CL interface had been introduced, which are thought to represent heterogeneous components, had disappeared in 4ch_1,2 006 and 4ch_2,1 008 into which mutations for regulating the VH/VL interface had been introduced, it became apparent that only target components can be efficiently prepared by applying regulation of the VH/VL interface in addition to regulation of the CH1/CL interface (FIGS. 10 and 11). In addition, the peaks which are thought to represent new heterogeneous components were not detected even if one further made mutations for regulating the VH/VL interface in 4ch_004 and 4ch_010, which had mutations introduced into CH1/CL only, and in which only components that were considered to be target components were thought to be purified (FIGS. 10 and 11).

From the above, it became apparent that the addition of regulation of the VH/VL interface to regulation of the CH1/CL interface further facilitates purification of target components, while it does not have a detrimental effect on the purification when only the target components are thought to be purified already.

[Example 6] Measurement of the Tm of Antibodies with Combined Modification Sites

Modifications for regulating the CH1/CL interface may have an effect on Fab stability. Therefore, Fab stability or Tm was measured in accordance with the method of Reference Example 2, for the combinations of TH2/TL17, TH13/TL16, and TH15/TL19. Antibodies in which the H chain/L chain consisted of TH2/TL17, TH13/TL16, and TH15/TL19 were prepared using an anti-IL6R antibody. The combinations of antibody modifications and the expressed antibodies are summarized in Table 11.

TABLE 11 Hch Lch SEQ SEQ name mutation ID NO name mutation ID NO Tm G1d  1 k0 13 95.0 TH2 Q175K  3 TL17 T180E, S131E, Q160E 30 93.1 TH13 K147E, Q175E 10 TL16 T180K, S131K, Q160K 29 95.1 TH15 K147E, Q175E, K213E 12 TL19 T180K, S131K, Q160K, E123K 32 94.8

Expression of antibodies was measured in accordance with Reference Example 1, and Tm (° C.) of each of the prepared antibodies was measured in accordance with Reference Example 2. The result shows that the values of Tm for the Fab of G1d/k0 which had no introduction of mutations, and for the Fab of TH2/TL17, TH13/TL16, and TH15/TL19, into which mutations were introduced into CH1/CL, were 95.0° C., 93.1° C., 95.1° C., and 94.8° C., respectively. It revealed that mutations for regulating the CH1/CL interface do not have an effect on Fab stability.

[Example 7] Effect of Introducing Mutations for Regulating the CH1/CL Interface on Binding Activity

The possibility of modifications for regulating the CH1/CL interface having an effect on antigen binding cannot be completely ruled out. Therefore, in order to measure the affinity for IL-6R and GPC3, binding activities of TH2/TL17, TH13/TL16, and TH15/TL19 were measured in accordance with the method of Reference Example 5 (Table 12).

Since the IL-6R-binding activity and GPC3-binding activity of TH2/TL17, TH13/TL16, and TH15/TL19, into which mutations for regulating the CH1/CL interface have been introduced, were not different from the binding activities of native Gld/k0 to IL-6R and GPC3, it became apparent that modifications for regulating the CH1/CL interface do not affect the affinities. Moreover, when the affinities for the two antigens, IL-6R and GPC3, were measured in accordance with Reference Example 5 using 4ch_004, 4ch_006, 4ch_008, and 4ch_010 prepared in Example 4, it was found that their affinities are equal to that of native Gld/k0 shown in Table 12 (Tables 13 and 14).

According to the studies conducted in Examples 1 to 7, it became apparent that only target components could be efficiently purified by introducing a mutation into CH1/CL, without lowering Fab stability and without lowering binding activity.

TABLE 12 Affinity to GPC3 Hch Lch name mutation SEQ ID NO name mutation SEQ ID NO ka kd KD(nM) G1d SEQ ID NO: 001 k0 SEQ ID NO: 013 2.7E+05 3.6E−04 1.3E−09 TH2 Q175K SEQ ID NO: 003 TL17 T180E, S131E, SEQ ID NO: 030 2.8E+05 3.9E−04 1.4E−09 Q160E TH13 K147E, Q175E SEQ ID NO: 010 TL16 T180K, S131K, SEQ ID NO: 029 2.7E+05 4.0E−04 1.5E−09 Q160K TH15 K147E, Q175E, SEQ ID NO: 012 TL19 T180K, S131K, SEQ ID NO: 032 3.9E+05 3.8E−04 9.9E−10 K213E Q160K, E123K Affinity to IL6R Hch Lch name mutation SEQ ID NO name mutation SEQ ID NO KLon Koff KD(nM) G1d SEQ ID NO: 001 k0 SEQ ID NO: 013 1.5E+05 4.1E−04 2.8E−09 TH2 Q175K SEQ ID NO: 003 TL17 T180E, S131E, SEQ ID NO: 030 1.3E+05 5.0E−04 3.8E−09 Q160E TH13 K147E, Q175E SEQ ID NO: 010 TL16 T180K, S131K, SEQ ID NO: 029 1.6E+05 4.4E−04 2.9E−09 Q160K TH15 K147E, Q175E, SEQ ID NO: 012 TL19 T180K, S131K, SEQ ID NO: 032 2.1E+05 4.8E−04 2.3E−09 K213E Q160K, E123K

TABLE 13 Ach Bch Affinity to GPC3 Affinity to IL6R NAME VH CH VL CL VH CH VL CL ka kd KD(nM) ka kd KD(nM) 4ch_001 MH0 G1d ML0 k0 GpH7 G1d GpL16 k0 2.7E+05 3.6E−04 1.3E−09 1 5E+05 4.1E−04 2.6E−09 4ch_004 MH0 TH2k ML0 TL17 GpH7 TH13h GpL16 TL16 2.7E+05 3.8E−04 1.4E−09 1.7E+05 4.7E−04 2.8E−09 4ch_006 MH0 TH2k ML0 TL17 GpH7 TH15h GpL16 TL19 3.4E+05 4.2E−04 1.2E−09 2.0E+05 4.3E−04 2.2E−09 4ch_001 MH0 G1d ML0 k0 GpH7 G1d GpL16 k0 2.7E+05 3.6E−04 1.3E−09 1.5E+05 4.1E−04 2.8E−09 4ch_008 MH0 TH13k ML0 TL16 GpH7 TH2h GpL16 TL17 2.6E+05 3.9E−04 1.5E−09 1.6E+05 4.6E−04 2.8E−09 4ch_010 MH0 TH15k ML0 TL19 GpH7 TH2h GpL16 TL17 2.9E+05 4.1E−04 1.4E−09 2.4E+05 5.9E−04 2.4E−09

TABLE 14 VH CH NAME chain name mutation SEQ ID NO name mutation SEQ ID NO 4ch_001 Ach MH0 SEQ ID NO: 036 G1d K147K SEQ ID NO: 001 4ch_004 Ach MH0 SEQ ID NO: 036 TH2k Q175K SEQ ID NO: 039 4ch_006 Ach MH0 SEQ ID NO: 036 TH2k Q175K SEQ ID NO: 039 4ch_001 Ach MH0 SEQ ID NO: 036 G1d K147K SEQ ID NO: 001 4ch_008 Ach MH0 SEQ ID NO: 036 TH13k K147E, Q175E SEQ ID NO: 040 4ch_010 Ach MH0 SEQ ID NO: 036 TH15k K147E, Q175E, SEQ ID NO: 041 K213E 4ch_001 Bch GpH7 SEQ ID NO: 034 G1d K147K SEQ ID NO: 001 4ch_004 Bch GpH7 SEQ ID NO: 034 TH13h K147E, Q175E SEQ ID NO: 044 4ch_006 Bch GpH7 SEQ ID NO: 034 TH15h K147E, Q175E, SEQ ID NO: 045 K213E 4ch_001 Bch GpH7 SEQ ID NO: 034 G1d K147K SEQ ID NO: 001 4ch_008 Bch GpH7 SEQ ID NO: 034 TH2h Q175K SEQ ID NO: 043 4ch_010 Bch GpH7 SEQ ID NO: 034 TH2h Q175K SEQ ID NO: 043 VL CL NAME chain name mutation SEQ ID NO name mutation SEQ ID NO 4ch_001 Ach ML0 SEQ ID NO: 037 k0 SEQ ID NO: 013 4ch_004 Ach ML0 SEQ ID NO: 037 TL17 T180E, S131E, SEQ ID NO: 030 Q160E 4ch_006 Ach ML0 SEQ ID NO: 037 TL17 T180E, S131E, SEQ ID NO: 030 Q160E 4ch_001 Ach ML0 SEQ ID NO: 037 k0 SEQ ID NO: 013 4ch_008 Ach ML0 SEQ ID NO: 037 TL16 T180K, S131K, SEQ ID NO: 029 Q160K 4ch_010 Ach ML0 SEQ ID NO: 037 TL19 T180K, S131K, SEQ ID NO: 032 Q160K, E123K 4ch_001 Bch GpL16 SEQ ID NO: 035 k0 SEQ ID NO: 013 4ch_004 Bch GpL16 SEQ ID NO: 035 TL16 T180K, S131K, SEQ ID NO: 029 Q160K 4ch_006 Bch GpL16 SEQ ID NO: 035 TL19 T180K, S131K, SEQ ID NO: 032 Q160K, E123K 4ch_001 Bch GpL16 SEQ ID NO: 035 k0 SEQ ID NO: 013 4ch_008 Bch GpL16 SEQ ID NO: 035 TL17 T180E, S131E, SEQ ID NO: 030 Q160E 4ch_010 Bch GpL16 SEQ ID NO: 035 TL17 T180E, S131E, SEQ ID NO: 030 Q160E

Example 8

Amino acid sequences of human IgA1 (SEQ ID NO: 63), IgA2 (SEQ ID NO: 64), IgD (SEQ ID NO: 65), IgE (SEQ ID NO: 66), IgG1 (SEQ ID NO: 67), IgG2 (SEQ ID NO: 68), IgG3 (SEQ ID NO: 69), IgG4 (SEQ ID NO: 70), and IgM (SEQ ID NO: 71) were aligned with respect to CH1 of the H chain; and amino acid sequences of human IgK (Kappa) (SEQ ID NO: 72), IgL1 (SEQ ID NO: 73), IgL2 (SEQ ID NO: 74), IgL3 (SEQ ID NO: 75), IgL6 (SEQ ID NO: 76), and IgL7 (SEQ ID NO: 77) (Lambda) were aligned with respect to CL of the L chain, followed by their respective comparisons. The results are shown in FIG. 12. Modifications discovered in the present example are indicated with arrows. As a result of introducing amino acids having different charges into the H chain and L chain so that the amino acids indicated with the arrows repel between CH1 of the H chain and CL of the L chain as indicated in the present example, it is thought that the target H chain and L chain can be specifically associated.

[Reference Example 1] Preparation of Antibody Expression Vectors, and Expression and Purification of Antibodies

Amino acid substitutions were introduced according to a method known among those skilled in the art using the QuikChange Site-Directed Mutagenesis Kit (Stratagene), PCR or the In-fusion Advantage PCR Cloning Kit (Takara), etc., followed by construction of expression vectors. The base sequences of the obtained expression vectors were determined according to a method known among those skilled in the art. Antibodies were expressed by transiently transfecting the prepared plasmids into human embryonic kidney cancer cell-derived HEK293H cell lines (Invitrogen) or FreeStyle 293 cells (Invitrogen). Antibodies were purified from the obtained culture supernatant according to a method known among those skilled in the art using rProtein A Sepharose™ Fast Flow (GE Healthcare). The concentration of purified antibodies was determined by measuring absorbance at 280 nm using a spectrophotometer, and the antibody concentration was calculated from the obtained value using an absorption coefficient calculated according to the PACE method (Protein Science 1995; 4: 2411-2423).

[Reference Example 2] Evaluation of the Melting Temperature (Tm) of Modified Antibodies by Differential Scanning Calorimetry

In this study, thermal stability was evaluated by measuring the melting temperature (Tm) of antibodies using a differential scanning calorimeter; MicroCal Capillary DSC (DKSH).

500 μL aliquot of each antibody solution was placed in a measuring plate and the temperature was increased from 20° C. to 115° C. The rate of temperature increase 120° C./hour and the change in heat capacity was monitored.

The data was analyzed using Origin? (Light Stone), and the temperature at which a change in heat capacity was observed was calculated and defined as the value of Tm.

[Reference Example 3] Anion Exchange Chromatography (AIEX) Analysis

Prepared antibodies were analyzed by the AIEX method using the Alliance System (Waters). Analyses were carried out according to the two-liquid gradient method using TSK-gel DEAE-NPR (Tosoh) for the analytical column, 10 mmol/L Tris-HCl (pH 7.5) for mobile phase A, and 10 mmol/L Tris-HCl and 500 mmol/L NaCl (pH 7.5) for mobile phase B. Measurements were carried out at a wavelength of 280 nm.

Data was analyzed using Empower2 (Waters), followed by calculation of the ratio of each detected peak.

[Reference Example 4] Cation Exchange Chromatography (CIEX) Analysis

Prepared antibodies were analyzed by the CIEX method using the Alliance System (Waters). Analyses were carried out according to the two-liquid gradient method using WCX-10 (Dionex) for the analytical column, 25 mmol/L MES (pH 6.1) for mobile phase A, and 25 mmol/L MES and 500 mmol/L NaCl (pH 6.1) for mobile phase B. Measurements were carried out at a wavelength of 280 nm.

Data was analyzed using Empower2 (Waters), followed by calculation of the ratio of each detected peak.

[Reference Example 5] Measurement of Affinity for IL6R and GPC3

Interactions between a target antibody and hIL6R or GPC3 were analyzed using Biacore T100 (GE Healthcare). HBS-EP+(GE Healthcare) was used for the running buffer, and the measurement temperature was 25° C. Protein A/G (Thermo Scientific) was immobilized on the Series S Sensor Chip CMS (GE Healthcare) by amine coupling, and was used as a chip. A target antibody was captured onto the chip, and interacted with each antigen diluted with running buffer. Antibodies captured on the chip were washed off by reacting with 10 mM glycine-HCl (pH 1.5) to regenerate the chip to be used repeatedly.

The dissociation constant (KD) of each antibody for antigen was calculated by carrying out kinetic analysis on the results of Biacore measurement. More specifically, the association rate constant ka (L/mol/s) and dissociation rate constant kd (1/s) were calculated by global fitting sensorgrams obtained by measuring with the Biacore Evaluation Software in a 1:1 Langmuir binding model, followed by calculation of dissociation constant KD (mol/L) from those values.

INDUSTRIAL APPLICABILITY

The method provided by the present invention enables one to regulate association without altering the structure, function, activity and the like of the original polypeptide (antibody), and is extremely useful since it requires only a small number of amino acid substitutions. In addition, the method also has little influence on antigenicity.

Use of the present invention enables efficient acquisition of bispecific antibodies that actually retain activity.

Claims

1. An antigen-binding molecule in which association of the heavy chain and light chain is regulated, wherein (a) the amino acid residue comprised in the heavy chain constant region (CH1) at position 147 as indicated by EU numbering, and the amino acid residue comprised in the light chain constant region (CL) at position 180 as indicated by EU numbering; (b) the amino acid residue comprised in CH1 at position 147 as indicated by EU numbering, and the amino acid residue comprised in CL at position 131 as indicated by EU numbering; and, (c) the amino acid residue comprised in CH1 at position 175 as indicated by EU numbering, and the amino acid residue comprised in CL at position 160 as indicated by EU numbering.

one set or two or more sets of amino acid residues selected from the group consisting of the sets of amino acid residues shown in (a) to (c) below in the heavy chain and light chain in the antigen-binding molecule are amino acid residues that mutually repel electrically:

2. The antigen-binding molecule of claim 1, further wherein amino acid residues in the set of amino acid residues shown in (d) below are amino acid residues that mutually repel electrically: (d) the amino acid residue comprised in CH1 at position 213 as indicated by EU numbering, and the amino acid residue comprised in CL at position 123 as indicated by EU numbering.

3. The antigen-binding molecule of claim 1 or 2, wherein the amino acid residues that mutually repel electrically are selected from amino acid residues comprised in either set of (X) and (Y) below:

(X) glutamic acid (E) or aspartic acid (D); and
(Y) lysine (K), arginine (R) or histidine (H).

4. The antigen-binding molecule of any one of claims 1 to 3, further wherein two or more amino acid residues forming an interface between the heavy chain variable region and light chain variable region are amino acid residues that mutually repel electrically.

5. The antigen-binding molecule of claim 4, wherein the amino acid residues that mutually repel electrically are one set or two sets of amino acid residues selected from the group consisting of the sets of amino acid residues shown in (a) or (b):

(a) the amino acid residue comprised in the heavy chain variable region at position 39 as indicated by Kabat numbering and the amino acid residue comprised in the light chain variable region at position 38 as indicated by Kabat numbering; or
(b) the amino acid residue comprised in the heavy chain variable region at position 45 as indicated by Kabat numbering, and the amino acid residue comprised in the light chain variable region at position 44 as indicated by Kabat numbering.

6. The antigen-binding molecule of claim 4 or 5, wherein the amino acid residues that mutually repel electrically are selected from the amino acid residues comprised in either set of (X) and (Y) below:

(X) glutamic acid (E) or aspartic acid (D); and
(Y) lysine (K), arginine (R), or histidine (H).

7. An antigen-binding molecule in which association of the heavy chain and light chain is regulated, wherein (a) the amino acid residue comprised in the heavy chain constant region (CH1) at position 147 as indicated by EU numbering, and the amino acid residue comprised in the light chain constant region (CL) at position 180 as indicated by EU numbering; (b) the amino acid residue comprised in CH1 at position 147 as indicated by EU numbering, and the amino acid residue comprised in CL at position 131 as indicated by EU numbering; and, (c) the amino acid residue comprised in CH1 at position 175 as indicated by EU numbering, and the amino acid residue comprised in CL at position 160 as indicated by EU numbering.

one set or two or more sets of amino acid residues selected from the group consisting of the sets of amino acid residues shown in (a) to (c) below in the associating heavy chain and light chain in the antigen-binding molecule are amino acid residues that do not mutually repel electrically:

8. The antigen-binding molecule of claim 7, further wherein amino acid residues of the set of amino acid residues shown in (d) below are amino acid residues that do not mutually repel electrically: (d) the amino acid residue comprised in CH1 at position 213 as indicated by EU numbering, and the amino acid residue comprised in CL at position 123 as indicated by EU numbering.

9. The antigen-binding molecule of claim 7 or 8, wherein the amino acid residues that do not mutually repel electrically are amino acid residues selected from each of two sets selected from the group consisting of (X) to (Z) below, and wherein the two sets are selected from among the combinations of (X) and (Y), (X) and (Z), (Y) and (Z), and (Z) and (Z):

(X) glutamic acid (E) or aspartic acid (D);
(Y) lysine (K), arginine (R) or histidine (H); and
(Z) alanine (A), asparagine (N), cysteine (C), glutamine (Q), glycine (G), isoleucine (I), leucine (L), methionine (M), phenylalanine (F), proline (P), serine (S), threonine (T), tryptophan (W), tyrosine (Y), or valine (V).

10. The antigen-binding molecule of any one of claims 7 to 9, wherein the amino acid residues that do not mutually repel electrically are the amino acid residue comprised in CH1 at position 175 as indicated by EU numbering which is lysine (K); and the amino acid residues comprised in CL at position 180, position 131, and position 160 as indicated by EU numbering which are all glutamic acid (E).

11. The antigen-binding molecule of any one of claims 7 to 9, wherein the amino acid residues that do not mutually repel electrically are the amino acid residues comprised in CH1 at position 147 and position 175 as indicated by EU numbering which are glutamic acid (E); and the amino acid residues comprised in CL at position 180, position 131, and position 160 as indicated by EU numbering which are all lysine (K).

12. The antigen-binding molecule of claim 11, further wherein the amino acid residue comprised in CH1 at position 213 as indicated by EU numbering is glutamic acid (E), and the amino acid residue comprised in CL at position 123 as indicated by EU numbering is lysine (K).

13. The antigen-binding molecule of any one of claims 7 to 12, wherein two or more amino acid residues forming the interface between the heavy chain variable region and light chain variable region are amino acid residues that do not mutually repel electrically.

14. The antigen-binding molecule of claim 13, wherein the amino acid residues that do not mutually repel electrically are one set or two sets of amino acid residues selected from the group consisting of the sets of amino acid residues shown in (a) or (b) below:

(a) the amino acid residue comprised in the heavy chain variable region at position 39 as indicated by Kabat numbering, and the amino acid residue comprised in the light chain variable region at position 38 as indicated by Kabat numbering; or
(b) the amino acid residue comprised in the heavy chain variable region at position 45 as indicated by Kabat numbering, and the amino acid residue comprised in the light chain variable region at position 44 as indicated by Kabat numbering.

15. The antigen-binding molecule of claim 13 or 14, wherein the amino acid residues that do not mutually repel electrically are amino acid residues selected from each of two sets selected from the group consisting of (X) to (Z) below, and wherein the two sets are selected from among the combinations of (X) and (Y), (X) and (Z), (Y) and (Z), and (Z) and (Z):

(X) glutamic acid (E) or aspartic acid (D);
(Y) lysine (K), arginine (R) or histidine (H); and
(Z) alanine (A), asparagine (N), cysteine (C), glutamine (Q), glycine (G), isoleucine (I), leucine (L), methionine (M), phenylalanine (F), proline (P), serine (S), threonine (T), tryptophan (W), tyrosine (Y), or valine (V).

16. The antigen-binding molecule of any one of claims 1 to 15, wherein the antigen-binding molecule is a bispecific antibody.

17. A method for producing an antigen-binding molecule in which association of the heavy chain and light chain is regulated, comprising steps of (1) to (3) below:

(1) modifying nucleic acids encoding the heavy chain constant region (CH1) and the light chain constant region (CL) so that one set or two or more sets of amino acid residues selected from the group consisting of the sets of amino acid residues shown in (a) to (c) below mutually repel electrically: (a) the amino acid residue comprised in CH1 at position 147 as indicated by EU numbering, and the amino acid residue comprised in CL at position 180 as indicated by EU numbering; (b) the amino acid residue comprised in CH1 at position 147 as indicated by EU numbering, and the amino acid residue comprised in CL at position 131 as indicated by EU numbering; and, (c) the amino acid residue comprised in CH1 at position 175 as indicated by EU numbering, and the amino acid residue comprised in CL at position 160 as indicated by EU numbering,
(2) introducing the modified nucleic acids into a host cell and culturing the host cell so that it expresses the nucleic acids, and
(3) collecting the antigen-binding molecule from a culture of the host cell.

18. The method for producing an antigen-binding molecule of claim 17, further comprising in step (1), modifying nucleic acids so that the amino acid residues in the set of amino acid residues shown in (d) below mutually repel electrically:

(d) the amino acid residue comprised in CH1 at position 213 as indicated by EU numbering, and the amino acid residue comprised in CL at position 123 as indicated by EU numbering.

19. The method for producing an antigen-binding molecule of claim 17 or 18, comprising in step (1), modifying nucleic acids so that the amino acid residues that mutually repel electrically are selected from among amino acid residues comprised in either group of (X) and (Y) below:

(X) glutamic acid (E) or aspartic acid (D); and
(Y) lysine (K), arginine (R), or histidine (H).

20. The method for producing an antigen-binding molecule of any one of claims 17 to 19, further comprising in step (1), modifying nucleic acids so that two or more amino acid residues forming the interface between the heavy chain variable region and light chain variable region are amino acid residues that mutually repel electrically.

21. The method for producing an antigen-binding molecule of claim 20, wherein the amino acid residues that mutually repel electrically are amino acid residues of any one set selected from the group consisting of the sets of amino acid residues shown in (a) or (b) below:

(a) the amino acid residue comprised in the heavy chain variable region at position 39 as indicated by Kabat numbering, and the amino acid residue comprised in the light chain variable region at position 38 as indicated by Kabat numbering; or
(b) the amino acid residue comprised in the heavy chain variable region at position 45 as indicated by Kabat numbering, and the amino acid residue comprised in the light chain variable region at position 44 as indicated by Kabat numbering.

22. The method for producing an antigen-binding molecule of claim 20 or 21, wherein the amino acid residues that mutually repel electrically are selected from amino acid residues comprised in either set of (X) and (Y) below:

(X) glutamic acid (E) or aspartic acid (D); and
(Y) lysine (K), arginine (R), or histidine (H).

23. A method for producing an antigen-binding molecule in which association of the heavy chain and light chain is regulated, comprising the following steps of (1) to (3):

(1) modifying nucleic acids encoding a heavy chain constant region (CH1) and a light chain constant region (CL) which associate so that one set or two or more sets of amino acid residues selected from the group consisting of the sets of amino acid residues shown in (a) to (c) below do not mutually repel electrically: (a) the amino acid residue comprised in the heavy chain constant region (CH1) at position 147 as indicated by EU numbering, and the amino acid residue comprised in the light chain constant region (CL) at position 180 as indicated by EU numbering; (b) the amino acid residue comprised in CH1 at position 147 as indicated by EU numbering, and the amino acid residue comprised in CL at position 131 as indicated by EU numbering; and, (c) the amino acid residue comprised in CH1 at position 175 as indicated by EU numbering, and the amino acid residue comprised in CL at position 160 as indicated by EU numbering,
(2) introducing the modified nucleic acids into a host cell and culturing the host cell so that it expresses the nucleic acids, and
(3) collecting the antigen-binding molecule from a culture of the host cell.

24. The method for producing an antigen-binding molecule of claim 23, further comprising in step (1), modifying nucleic acids so that the amino acid residues in the set of amino acid residues shown in (d) below do not mutually repel electrically:

(d) the amino acid residue comprised in CH1 at position 213 as indicated by EU numbering, and the amino acid residue comprised in CL at position 123 as indicated by EU numbering.

25. The method for producing an antigen-binding molecule of claim 23 or 24, comprising in step (1), modifying the nucleic acids so that the amino acid residues that do not mutually repel electrically are amino acids residues selected from each of two sets selected from the group consisting of (X) to (Z) below, and wherein the two sets are selected from among the combinations of (X) and (Y), (X) and (Z), (Y) and (Z), and (Z) and (Z):

(X) glutamic acid (E) or aspartic acid (D);
(Y) lysine (K), arginine (R), or histidine (H); and
(Z) alanine (A), asparagine (N), cysteine (C), glutamine (Q), glycine (G), isoleucine (I), leucine (L), methionine (M), phenylalanine (F), proline (P), serine (S), threonine (T), tryptophan (W), tyrosine (Y), or valine (V).

26. The method for producing an antigen-binding molecule of any one of claims 23 to 25, comprising in step (1), modifying nucleic acids so that the amino acid residues that do not mutually repel electrically are the amino acid residue comprised in CH1 at position 175 as indicated by EU numbering which is lysine (K), and the amino acid residues comprised in CL at position 180, position 131, and position 160 as indicated by EU numbering which are all glutamic acid (E).

27. The method for producing an antigen-binding molecule of any one of claims 23 to 25, comprising in step (1), modifying nucleic acids so that the amino acid residues that do not mutually repel electrically are the amino acid residues comprised in CH1 at position 147 and position 175 as indicated by EU numbering which are glutamic acid (E), and the amino acid residues comprised in CL at position 180, position 131, and position 160 as indicated by EU numbering which are all lysine (K).

28. The method for producing an antigen-binding molecule of claim 27, further comprising modifying nucleic acids so that the amino acid residue comprised in CH1 at position 213 as indicated by EU numbering is glutamic acid (E), and the amino acid residue comprised in CL at position 123 as indicated by EU numbering is lysine (K).

29. The method for producing an antigen-binding molecule of any one of claims 23 to 28, further comprising in step (1), modifying nucleic acids so that two or more amino acid residues forming the interface between the heavy chain variable region and light chain variable region are amino acid residues that do not mutually repel electrically.

30. The method for producing an antigen-binding molecule of claim 29, wherein the amino acid residues that do not mutually repel electrically are amino acid residues of any one set selected from the group consisting of the sets of amino acid residues shown in (a) or (b) below:

(a) the amino acid residue comprised in the heavy chain variable region at position 39 as indicated by Kabat numbering, and the amino acid residue comprised in the light chain variable region at position 38 as indicated by Kabat numbering; or
(b) the amino acid residue comprised in the heavy chain variable region at position 45 as indicated by Kabat numbering, and the amino acid residue comprised in the light chain variable region at position 44 as indicated by Kabat numbering.

31. The method for producing an antigen-binding molecule of claim 29 or 30, wherein the amino acid residues that do not mutually repel electrically are amino acid residues selected from each of two sets selected from the group consisting of (X) to (Z) below, and wherein the two sets are selected from among the combinations of (X) and (Y), (X) and (Z), (Y) and (Z), and (Z) and (Z):

(X) glutamic acid (E) or aspartic acid (D);
(Y) lysine (K), arginine (R) or histidine (H); and
(Z) alanine (A), asparagine (N), cysteine (C), glutamine (Q), glycine (G), isoleucine (I), leucine (L), methionine (M), phenylalanine (F), proline (P), serine (S), threonine (T), tryptophan (W), tyrosine (Y), or valine (V).

32. An antigen-binding molecule produced according to the method for producing an antigen-binding molecule of any one of claims 17 to 31.

33. The antigen-binding molecule of claim 32, wherein the antigen-binding molecule is a bispecific antibody.

34. A method for regulating association of the heavy chain and light chain of an antigen-binding molecule, comprising: (a) the amino acid residue comprised in CH1 at position 147 as indicated by EU numbering, and the amino acid residue comprised in CL at position 180 as indicated by EU numbering; (b) the amino acid residue comprised in CH1 at position 147 as indicated by EU numbering, and the amino acid residue comprised in CL at position 131 as indicated by EU numbering; and (c) the amino acid residue comprised in CH1 at position 175 as indicated by EU numbering, and the amino acid residue comprised in CL at position 160 as indicated by EU numbering.

modifying nucleic acids so that one set or two or more sets of amino acid residues selected from the group consisting of the sets of amino acid residues shown in (a) to (c) below are amino acid residues that mutually repel electrically:

35. The method of claim 34, further comprising modifying nucleic acids so that the amino acid residues in the set of amino acid residues shown in (d) below are amino acid residues that mutually repel electrically: (d) the amino acid residue comprised in CH1 at position 213 as indicated by EU numbering, and the amino acid residue comprised in CL at position 123 as indicated by EU numbering.

36. The method of claim 34 or 35, wherein the amino acid residues that mutually repel electrically are selected from amino acid residues comprised in either set of (X) and (Y) below:

(X) glutamic acid (E) or aspartic acid (D); and
(Y) lysine (K), arginine (R), or histidine (H).

37. The method of any one of claims 34 to 36, wherein two or more amino acid residues forming the interface between the heavy chain variable region and light chain variable region are amino acid residues that mutually repel electrically.

38. The method of claim 37, wherein the amino acid residues that mutually repel electrically are amino acid residues of any one set selected from the group consisting of the sets of amino acid residues shown in (a) or (b) below:

(a) the amino acid residue comprised in the heavy chain variable region at position 39 as indicated by Kabat numbering, and the amino acid residue comprised in the light chain variable region at position 38 as indicated by Kabat numbering; or
(b) the amino acid residue comprised in the heavy chain variable region at position 45 as indicated by Kabat numbering, and the amino acid residue comprised in the light chain variable region at position 44 as indicated by Kabat numbering.

39. The method of claim 37 or 38, wherein the amino acid residues that mutually repel electrically are selected from amino acid residues comprised in either set of (X) and (Y) below:

(X) glutamic acid (E) or aspartic acid (D); and
(Y) lysine (K), arginine (R), or histidine (H).

40. A method for regulating association of the heavy chain and light chain of an antigen-binding molecule, comprising: (a) the amino acid residue comprised in CH1 at position 147 as indicated by EU numbering, and the amino acid residue comprised in CL at position 180 as indicated by EU numbering; (b) the amino acid residue comprised in CH1 at position 147 as indicated by EU numbering, and the amino acid residue comprised in CL at position 131 as indicated by EU numbering; and, (c) the amino acid residue comprised in CH1 at position 175 as indicated by EU numbering, and the amino acid residue comprised in CL at position 160 as indicated by EU numbering.

modifying nucleic acids so that one set or two or more sets of amino acid residues selected from the group consisting of the sets of amino acid residues shown in (a) to (c) below in the associating heavy chain and light chain in the antigen-binding molecule are amino acid residues that do not mutually repel electrically:

41. The method of claim 40, further comprising modifying nucleic acids so that the amino acid residues in the set of amino acid residues shown in (d) below are amino acid residues that do not mutually repel electrically: (d) the amino acid residue comprised in CH1 at position 213 as indicated by EU numbering, and the amino acid residue comprised in CL at position 123 as indicated by EU numbering.

42. The method of claim 40 or 41, wherein the amino acid residues that do not mutually repel electrically are amino acid residues selected from each of two sets selected from the group consisting of (X) to (Z) below, and wherein the two sets are selected from among the combinations of (X) and (Y), (X) and (Z), (Y) and (Z), and (Z) and (Z):

(X) glutamic acid (E) or aspartic acid (D);
(Y) lysine (K), arginine (R), or histidine (H); and
(Z) alanine (A), asparagine (N), cysteine (C), glutamine (Q), glycine (G), isoleucine (I), leucine (L), methionine (M), phenylalanine (F), proline (P), serine (S), threonine (T), tryptophan (W), tyrosine (Y), or valine (V).

43. The method of any one of claims 40 to 42, wherein the amino acid residues that do not mutually repel electrically are the amino acid residue comprised in CH1 at position 175 as indicated by EU numbering which is lysine (K), and the amino acid residues comprised in CL at position 180, position 131, and position 160 as indicated by EU numbering which are all glutamic acid (E).

44. The method of any one of claims 40 to 42, wherein the amino acid residues that do not mutually repel electrically are the amino acid residues comprised in CH1 at position 147 and position 175 as indicated by EU numbering which are glutamic acid (E), and the amino acid residues comprised in CL at position 180, position 131, and position 160 as indicated by EU numbering which are all lysine (K).

45. The method of claim 44, wherein the amino acid residue comprised in CH1 at position 213 as indicated by EU numbering is glutamic acid (E), and the amino acid residue contained in CL at position 123 as indicated by EU numbering is lysine (K).

46. The method of any one of claims 34 to 45, wherein the antigen-binding molecule is a bispecific antibody.

47. A composition containing the antigen-binding molecule of any one of claims 1 to 16, 32, and 33, and a pharmaceutically acceptable carrier.

48. A nucleic acid encoding the antigen-binding molecule of any one of claims 1 to 16, 32, and 33.

49. A host cell having the nucleic acid of claim 48.

Patent History
Publication number: 20200087380
Type: Application
Filed: Nov 22, 2019
Publication Date: Mar 19, 2020
Applicant: Chugai Seiyaku Kabushiki Kaisha (Tokyo)
Inventors: Taichi Kuramochi (Shizuoka), Meiri Kawazoe (Shizuoka), Naoka Hironiwa (Shizuoka), Tomoyuki Igawa (Shizuoka)
Application Number: 16/692,676
Classifications
International Classification: C07K 16/00 (20060101); C07K 16/28 (20060101); C07K 16/30 (20060101); C07K 16/46 (20060101);