ANTIGEN-BINDING MOLECULE FOR ELIMINATING AGGREGATED ANTIGENS

Abstract
The present inventors discovered that incorporating an Fc region and an antigen-binding domain whose antigen-binding activity varies depending on ion concentration into an antigen-binding molecule that binds to an aggregate-forming antigen produces an antigen-binding molecule that can preferentially clear protein aggregates in comparison to protein monomers from plasma. Use of antigen-binding molecules of the present invention allows various diseases stemming from target tissues to be treated target-tissue-specifically. Use of antigen-binding molecules of the present invention enables treatment of diseases caused by protein aggregates.
Description
TECHNICAL FIELD

The present invention provides uses of antigen-binding molecules for eliminating aggregated antigens from plasma; methods for eliminating aggregated antigens from plasma, which comprise administering antigen-binding molecules; pharmaceutical compositions comprising antigen-binding molecules that are capable of eliminating aggregated antigens from plasma; methods of screening for antigen-binding molecules for eliminating aggregated antigens from plasma; and methods for producing antigen-binding molecules for eliminating aggregated antigens from plasma.


BACKGROUND ART

When proteins form aggregates due to various factors such as gene mutations and environmental changes, they are known to become causes of various diseases by reducing physiological functions of proteins or by posing toxic effects on cells. For example, when amyloid-β aggregates and accumulates in the brain, nerve cells degenerate and Alzheimer's disease develops. Furthermore, when immunoglobulin L chain aggregates and deposits in each organ to cause organ failure, AL amyloidosis develops. Similar to these diseases, a group of diseases characterized by extracellular accumulation of various protein aggregates is called amyloidosis, and the classification by The Research Committees on intractable Diseases Specified by the Japanese Ministry of Health and Welfare (now Japanese Ministry of Health, Labour and Welfare) reports that there are ten disease types of systemic amyloidosis and ten disease types of localized amyloidosis. Besides amyloidosis, α-synuclein disease is known as a disease where aggregated α-synuclein is deposited in nerve cells. Some inherited Parkinson's diseases are caused by deposition of α-synuclein in cerebral neurons, and they are considered to be a type of α-synuclein disease. As described above, many diseases caused by protein aggregates are known in the world, but the mechanism of protein aggregation is still unclear; and for many of these diseases, an ultimate therapeutic agent does not exist.


Recently, antibodies are drawing attention as pharmaceuticals as they have a high stability in plasma and have few side effects. At present, a number of IgG-type antibody pharmaceuticals are available on the market and many antibody pharmaceuticals are currently under development (Non-patent Documents 1 and 2).


Meanwhile, the antigen-neutralizing capacity of a single antibody molecule depends on its affinity. By increasing the affinity, an antigen can be neutralized by a smaller amount of an antibody. Various methods can be used to enhance antibody affinity (Non-patent Document 6). Furthermore, if the affinity could be made infinite by covalently binding the antibody to the antigen, a single antibody molecule could neutralize one antigen molecule (a divalent antibody can neutralize two antigen molecules). However, the stoichiometric neutralization of one antibody against one antigen (one divalent antibody against two antigens) is the limit of pre-existing methods, and thus it was impossible to completely neutralize antigen with an amount of antibody smaller than the amount of antigen. In other words, the affinity-enhancing effect has a limit (Non-patent Document 9). To prolong the neutralization effect of a neutralizing antibody for a certain period, the antibody must be administered at a dose higher than the amount of antigen produced in the body during the same period. Therefore, with just the above-described improvement of antibody pharmacokinetics or affinity maturation technology, there were limitations when it comes to reduction of the required antibody dose. Accordingly, in order to sustain antibody's antigen-neutralizing effect for a target period with an amount of the antibody smaller than the amount of antigen, a single antibody must neutralize multiple antigens.


As a novel method for achieving this objective, use of an antibody that binds to an antigen in a pH-dependent manner has been reported recently to enable a single antibody molecule to bind to multiple antigen molecules (Patent Document 1 and Non-patent Document 5). Antibodies with pH-dependent antigen binding, which bind strongly to an antigen under the neutral condition in plasma, and dissociate from the antigen under the acidic condition in the endosome, can dissociate from the antigen in the endosome. When an antibody with pH-dependent antigen binding that has dissociated from the antigen is recycled into plasma by FcRn, the antibody can again bind to an antigen; therefore, a single pH-dependent antigen-binding antibody molecule can repeatedly bind to multiple antigens. Such a recycling antibody will be very useful as a pharmaceutical since a single antibody molecule can repeatedly bind to multiple antigens.


In addition, the plasma retention of an antigen is very short when compared to that of antibodies that are recycled by binding to FcRn. When a typical antibody with long plasma retention binds to such an antigen, the plasma retention of the antigen-antibody complex is prolonged to the same as that of the antibody. Thus, when a typical antibody is administered, the antibody binds to an antigen, the plasma retention of the antigen is prolonged (becomes difficult to be eliminated from plasma) by binding to the antibody, and thus the plasma antigen concentration is increased. On the other hand, antibodies with pH-dependent antigen binding can suppress increase of antigen concentration in plasma by dissociating from the antigen in the endosome. However, even with such a pH-dependent antigen-binding antibody, the plasma antigen concentration may be increased by antibody administration compared to before antibody administration.


Recently, antibodies were produced by enhancing the FcRn-binding property of antibodies with pH-dependent antigen binding under a neutral condition, and it was found that administration of such antibodies can decrease the antigen concentration in plasma compared to before antibody administration (Patent Document 2). While antibody administration of recycled antibodies such as antibodies with pH-dependent antigen binding and typical antibodies result in an increase of antigen concentration in plasma, pH-dependent antigen-binding antibodies with enhanced FcRn-binding under a neutral condition can decrease antigen concentration in plasma by antibody administration. Such antibodies are very useful as pharmaceuticals since they can actively eliminate antigens from plasma.


However, for diseases where protein aggregates is a cause of disease, monomers that have normal function co-exist in plasma with the disease-causing aggregates, and antibodies having a property of selectively eliminating aggregates from plasma are desired. Prior art documents of the present invention are shown below.


PRIOR ART DOCUMENTS
Patent Documents
[Patent Document 1] WO 2009/125825, ANTIGEN-BINDING MOLECULE CAPABLE OF BINDING TO TWO OR MORE ANTIGEN MOLECULES REPEATEDLY
[Patent Document 2] WO 2011/122011, ANTIBODIES WITH MODIFIED AFFINITY TO FCRN THAT PROMOTE ANTIGEN CLEARANCE
Non-Patent Documents

[Non-patent Document 1] Monoclonal antibody successes in the clinic, Janice M Reichert, Clark J Rosensweig, Laura B Faden & Matthew C Dewitz, Nature Biotechnology 23, 1073-1078 (2005)


[Non-patent Document 2] Pavlou A K, Belsey M J., The therapeutic antibodies market to 2008., Eur J Pharm Biopharm. 2005 April; 59(3): 389-96.


[Non-patent Document 3] Rajpal A, Beyaz N, Haber L, Cappuccilli G, Yee H, Bhatt R R, Takeuchi T, Lerner R A, Crea R., A general method for greatly improving the affinity of antibodies by using combinatorial libraries., Proc. Natl. Acad. Sci. U.S.A (2005) 102(24), 8466-8471


[Non-patent Document 4] Rathanaswami P, Roalstad S, Roskos L, Su Q J, Lackie S, Babcook J., Demonstration of an in vivo generated sub-picomolar affinity fully human monoclonal antibody to interleukin-8., Biochem. Biophys. Res. Commun. (2005) 334(4), 1004-1013


[Non-patent Document 5] Igawa T, et al., Antibody recycling by engineered pH-dependent antigen binding improves the duration of antigen neutralization. Nat Biotechnol. 2010, 28, 1203-7.


SUMMARY OF INVENTION
Problems to be Solved by the Invention

For treatment of diseases where protein aggregates is a cause of the disease, antibodies having a property of selectively eliminating aggregates from plasma are desired. However, typical antibodies of which FcRn-binding is not enhanced under neutral conditions may possibly increase antigen concentration in plasma as described in the Background Art. In such cases, elimination of proteins causing the disease is delayed, and such proteins will tend to accumulate, causing negative effects such as enhanced cytotoxicity.


On the other hand, while pH-dependent antigen-binding antibodies with enhanced FcRn binding can eliminate antigens from plasma, in diseases where aggregates is the cause, monomers having normal functions coexist in plasma with aggregates that cause the disease, and therefore even if a pH-dependent antigen-binding antibody with enhanced FcRn binding is used, not only the aggregates but also normal monomers may be eliminated as well. Furthermore, when the proportion of the monomers present in plasma is overwhelmingly large relative to the aggregates, there is also a possibility that elimination of the monomers might be carried out preferentially and that elimination of the aggregates may become difficult.


The present invention was made in view of such circumstances. An objective of the present invention is to provide antigen-binding molecules that can eliminate disease-causing protein aggregates in preference to protein monomers from plasma, pharmaceutical compositions comprising the antigen-binding molecules, and methods of producing the antigen-binding molecules.


Means for Solving the Problems

The present inventors conducted dedicated studies to solve the above-mentioned objectives. As a result, the present inventors successfully produced antigen-binding molecules that can eliminate protein aggregates in preference to monomers from plasma, by introducing an Fc region and an antigen-binding domain whose antigen-binding activity varies depending on ion concentration into an antigen-binding molecule that binds to an aggregate-forming antigen.


More specifically, the present invention relates to the following:


[1] an antigen-binding molecule which binds to an aggregated antigen, and comprises an Fc region and an antigen-binding domain whose antigen-binding activity varies depending on an ion concentration condition;


[2] the antigen-binding molecule of [1], wherein binding activity for the aggregated antigen is higher than binding activity for an unaggregated antigen;


[3] the antigen-binding molecule of [1] or [2], in which binding activity to an Fcγ receptor or an FcRn of a complex formed between an aggregated antigen and the antigen-binding molecule is higher than binding activity to an Fcγ receptor or an FcRn of a complex formed between an unaggregated antigen and the antigen-binding molecule;


[4] the antigen-binding molecule of any one of [1] to [3], which eliminates an aggregated antigen from plasma in preference to an unaggregated antigen;


[5] the antigen-binding molecule of any one of [1] to [4], wherein the ratio of plasma clearance of an aggregated antigen in the absence of the antigen-binding molecule to plasma clearance of an aggregated antigen in the presence of the antigen-binding molecule is 1.5 times or more than the same plasma clearance ratio for an unaggregated antigen;


[6] the antigen-binding molecule of any one of [1] to [5], wherein an antigen-binding activity of the antigen-binding domain varies depending on a calcium ion concentration condition;


[7] the antigen-binding molecule of [6], wherein the antigen-binding domain is an antigen-binding domain whose antigen-binding activity under a low calcium ion concentration condition is lower than its antigen-binding activity under a high calcium ion concentration condition;


[8] the antigen-binding molecule of any one of [1] to [7], wherein the antigen-binding domain is an antigen-binding domain whose antigen-binding activity varies depending on a pH condition;


[9] the antigen-binding molecule of [8], wherein the antigen-binding domain is an antigen-binding domain whose antigen-binding activity in an acidic pH range is lower than its antigen-binding activity in a neutral pH range condition;


[10] the antigen-binding molecule of any one of [1] to [9], wherein the antigen is an antigen that aggregates in plasma;


[11] the antigen-binding molecule of [10], wherein the antigen is huntingtin, ataxin-1, ataxin-2, Ca channel α1A, ataxin-7, TATA binding protein, MDJ, DRPLA, androgen receptor, α1-antitrypsin, α1-antichymotrypsin, neuroserpin, C1 inhibitor, antithrombin III, Aβ, L-ch, transthyretin, SAA, β2M, H-ch, cystatin C, α synuclein, amylin, hemoglobin, crystalline, IgA, Tau protein, TAR DNA-binding protein 43 kDa (TDP-43), Superoxide dismutase (SOD1), FUS (Fused in Sarcoma gene), Prion, PHOX2B, ARX, poly-adenylate binding protein nuclear 1 (PABPN1), dysferlin, desmin, GFAP, or keratin 5/14;


[12] the antigen-binding molecule of any one of [1] to [11], wherein the Fc region is an Fc region represented by any one of SEQ ID NO: 9, 10, 11, or 12;


[13] the antigen-binding molecule of any one of [1] to [11], wherein under an acidic pH condition, FcRn-binding activity of the Fc region is enhanced compared to that of the Fc region represented by any one of SEQ ID NO: 9, 10, 11, or 12;


[14] the antigen-binding molecule of [13], wherein the Fc region is an Fc region in which at least one or more amino acids selected from the group consisting of amino acids at positions 238, 244, 245, 249, 250, 251, 252, 253, 254, 255, 256, 257, 258, 260, 262, 265, 270, 272, 279, 283, 285, 286, 288, 293, 303, 305, 307, 308, 309, 311, 312, 314, 316, 317, 318, 332, 339, 340, 341, 343, 356, 360, 362, 375, 376, 377, 378, 380, 382, 385, 386, 387, 388, 389, 400, 413, 415, 423, 424, 427, 428, 430, 431, 433, 434, 435, 436, 438, 439, 440, 442, and 447, according to EU numbering, are substituted in the amino acid sequence of the Fc region represented by any one of SEQ ID NO: 9, 10, 11, or 12;


[15] the antigen-binding molecule of [14], wherein the Fc region comprises at least one or more amino acids selected from the group consisting of:


Leu for the amino acid at position 238;


Leu for the amino acid at position 244;


Arg for the amino acid at position 245;


Pro for the amino acid at position 249;


Gln or Glu for the amino acid at position 250, or


Arg, Asp, Glu, or Leu for the amino acid at position 251;


Phe, Ser, Thr, or Tyr for the amino acid at position 252;


Ser or Thr for the amino acid at position 254;


Arg, Gly, Ile, or Leu for the amino acid at position 255;


Ala, Arg, Asn, Asp, Gln, Glu, Pro, or Thr for the amino acid at position 256;


Ala, Ile, Met, Asn, Ser, or Val for the amino acid at position 257;


Asp for the amino acid at position 258;


Ser for the amino acid at position 260;


Leu for the amino acid at position 262;


Lys for the amino acid at position 270;


Leu or Arg for the amino acid at position 272;


Ala, Asp, Gly, His, Met, Asn, Gln, Arg, Ser, Thr, Trp, or Tyr for the amino acid at position 279;


Ala, Asp, Phe, Gly, His, Ile, Lys, Leu, Asn, Pro, Gln, Arg, Ser, Thr, Trp, or Tyr for the amino acid at position 283;


Asn for the amino acid at position 285;


Phe for the amino acid at position 286;


Asn or Pro for the amino acid at position 288;


Val for the amino acid at position 293;


Ala, Glu, Gln, or Met for the amino acid at position 307;


Ala, Glu, Ile, Lys, Leu, Met, Ser, Val, or Trp for the amino acid at position 311;


Pro for the amino acid at position 309;


Ala, Asp, or Pro for the amino acid at position 312;


Ala or Leu for the amino acid at position 314;


Lys for the amino acid at position 316;


Pro for the amino acid at position 317;


Asn or Thr for the amino acid at position 318;


Phe, His, Lys, Leu, Met, Arg, Ser, or Trp for the amino acid at position 332;


Asn, Thr, or Trp for the amino acid at position 339;


Pro for the amino acid at position 341;


Glu, His, Lys, Gln, Arg, Thr, or Tyr for the amino acid at position 343;


Arg for the amino acid at position 375;


Gly, Ile, Met, Pro, Thr, or Val for the amino acid at position 376;


Lys for the amino acid at position 377;


Asp, Asn, or Val for the amino acid at position 378;


Ala, Asn, Ser, or Thr for the amino acid at position 380;


Phe, His, Ile, Lys, Leu, Met, Asn, Gln, Arg, Ser, Thr, Val, Trp, or Tyr for the amino acid at position 382;


Ala, Arg, Asp, Gly, His, Lys, Ser, or Thr for the amino acid at position 385;


Arg, Asp, Ile, Lys, Met, Pro, Ser, or Thr for the amino acid at position 386;


Ala, Arg, His, Pro, Ser, or Thr for the amino acid at position 387;


Asn, Pro, or Ser for the amino acid at position 389;


Asn for the amino acid at position 423;


Asn for the amino acid at position 427;


Leu, Met, Phe, Ser, or Thr for the amino acid at position 428;


Ala, Phe, Gly, His, Ile, Lys, Leu, Met, Asn, Gln, Arg, Ser, Thr, Val, or Tyr for the amino acid at position 430;


His or Asn for the amino acid at position 431;


Arg, Gln, His, Ile, Lys, Pro, or Ser for the amino acid at position 433;


Ala, Gly, His, Phe, Ser, Trp, or Tyr for the amino acid at position 434;


Arg, Asn, His, Ile, Leu, Lys, Met, or Thr for the amino acid at position 436;


Lys, Leu, Thr, or Trp for the amino acid at position 438;


Lys for the amino acid at position 440, or


Lys for the amino acid at position 442; and


Ile, Pro, or Thr for the amino acid at position 308;


as indicated by EU numbering, in the amino acid sequence of the Fc region represented by any one of SEQ ID NO: 9, 10, 11, or 12;


[16] the antigen-binding molecule of any one of [1] to [11], wherein under a neutral pH range condition, an FcRn-binding activity of the Fc region is enhanced compared to that of the Fc region represented by any one of SEQ ID NO: 9, 10, 11, or 12;


[17] the antigen-binding molecule of [16], wherein the Fc region is an Fc region in which at least one or more amino acids selected from the group consisting of amino acids at positions 237, 248, 250, 252, 254, 255, 256, 257, 258, 265, 286, 289, 297, 298, 303, 305, 307, 308, 309, 311, 312, 314, 315, 317, 332, 334, 360, 376, 380, 382, 384, 385, 386, 387, 389, 424, 428, 433, 434, and 436, according to EU numbering, are substituted in the amino acid sequence of the Fc region represented by any one of SEQ ID NO: 9, 10, 11, or 12;


[18] the antigen-binding molecule of [17], wherein the Fc region comprises at least one or more amino acids selected from the group of:


Met for the amino acid at position 237;


Ile for the amino acid at position 248;


Ala, Phe, Ile, Met, Gln, Ser, Val, Trp, or Tyr for the amino acid at position 250;


Phe, Trp, or Tyr for the amino acid at position 252;


Thr for the amino acid at position 254;


Glu for the amino acid at position 255;


Asp, Asn, Glu, or Gln for the amino acid at position 256;


Ala, Gly, Ile, Leu, Met, Asn, Ser, Thr, or Val for the amino acid at position 257;


His for the amino acid at position 258;


Ala for the amino acid at position 265;


Ala or Glu for the amino acid at position 286;


His for the amino acid at position 289;


Ala for the amino acid at position 297;


Ala for the amino acid at position 303;


Ala for the amino acid at position 305;


Ala, Asp, Phe, Gly, His, Ile, Lys, Leu, Met, Asn, Pro, Gln, Arg, Ser, Val, Trp, or Tyr for the amino acid at position 307;


Ala, Phe, Ile, Leu, Met, Pro, Gln, or Thr for the amino acid at position 308;


Ala, Asp, Glu, Pro, or Arg for the amino acid at position 309;


Ala, His, or Ile for the amino acid at position 311;


Ala or His for the amino acid at position 312;


Lys or Arg for the amino acid at position 314;


Ala, Asp, or His for the amino acid at position 315;


Ala for the amino acid at position 317;


Val for the amino acid at position 332;


Leu for the amino acid at position 334;


His for the amino acid at position 360;


Ala for the amino acid at position 376;


Ala for the amino acid at position 380;


Ala for the amino acid at position 382;


Ala for the amino acid at position 384;


Asp or His for the amino acid at position 385;


Pro for the amino acid at position 386;


Glu for the amino acid at position 387;


Ala or Ser for the amino acid at position 389;


Ala for the amino acid at position 424;


Ala, Asp, Phe, Gly, His, Ile, Lys, Leu, Asn, Pro, Gln, Ser, Thr, Val, Trp, or Tyr for the amino acid at position 428;


Lys for the amino acid at position 433;


Ala, Phe, His, Ser, Trp, or Tyr for the amino acid at position 434; and


His, Ile, Leu, Phe, Thr, or Val for the amino acid at position 436;


as indicated by EU numbering in the amino acid sequence of the Fc region represented by any one of SEQ ID NOs: 9, 10, 11, and 12;


[19] the antigen-binding molecule of any one of [1] to [15], wherein the Fc region includes an Fc region that has a higher Fcγ receptor-binding activity than that of the Fc region of a native human IgG;


[20] the antigen-binding molecule of [19], wherein the Fc region comprises in its amino acid sequence at least one or more amino acids that are different from amino acids of the native human IgG Fc region selected from the group of positions 221, 222, 223, 224, 225, 227, 228, 230, 231, 232, 233, 234, 235, 236, 237, 238, 239, 240, 241, 243, 244, 245, 246, 247, 249, 250, 251, 254, 255, 256, 258, 260, 262, 263, 264, 265, 266, 267, 268, 269, 270, 271, 272, 273, 274, 275, 276, 278, 279, 280, 281, 282, 283, 284, 285, 286, 288, 290, 291, 292, 293, 294, 295, 296, 297, 298, 299, 300, 301, 302, 303, 304, 305, 311, 313, 315, 317, 318, 320, 322, 323, 324, 325, 326, 327, 328, 329, 330, 331, 332, 333, 334, 335, 336, 337, 339, 376, 377, 378, 379, 380, 382, 385, 392, 396, 421, 427, 428, 429, 434, 436, and 440 (EU numbering);


[21] the antigen-binding molecule of [20], wherein the Fc region comprises in its amino acid sequence at least one or more amino acid selected from the group of:


Lys or Tyr for the amino acid at position 221;


Phe, Trp, Glu, or Tyr for the amino acid at position 222;


Phe, Trp, Glu, or Lys for the amino acid at position 223;


Phe, Trp, Glu, or Tyr for the amino acid at position 224;


Glu, Lys, or Trp for the amino acid at position 225;


Glu, Gly, Lys, or Tyr for the amino acid at position 227;


Glu, Gly, Lys, or Tyr for the amino acid at position 228;


Ala, Glu, Gly, or Tyr for the amino acid at position 230;


Glu, Gly, Lys, Pro, or Tyr for the amino acid at position 231;


Glu, Gly, Lys, or Tyr for the amino acid at position 232;


Ala, Asp, Phe, Gly, His, Ile, Lys, Leu, Met, Asn, Gln, Arg, Ser, Thr, Val, Trp, or Tyr for the amino acid at position 233;


Ala, Asp, Glu, Phe, Gly, His, Ile, Lys, Met, Asn, Pro, Gln, Arg, Ser, Thr, Val, Trp, or Tyr for the amino acid at position 234;


Ala, Asp, Glu, Phe, Gly, His, Ile, Lys, Met, Asn, Pro, Gln, Arg, Ser, Thr, Val, Trp, or Tyr for the amino acid at position 235;


Ala, Asp, Glu, Phe, His, Ile, Lys, Leu, Met, Asn, Pro, Gln, Arg, Ser, Thr, Val, Trp, or Tyr for the amino acid at position 236;


Asp, Glu, Phe, His, Ile, Lys, Leu, Met, Asn, Pro, Gln, Arg, Ser, Thr, Val, Trp, or Tyr for the amino acid at position 237;


Asp, Glu, Phe, Gly, His, Ile, Lys, Leu, Met, Asn, Gln, Arg, Ser, Thr, Val, Trp, or Tyr for the amino acid at position 238;


Asp, Glu, Phe, Gly, His, Ile, Lys, Leu, Met, Asn, Pro, Gln, Arg, Thr, Val, Trp, or Tyr for the amino acid at position 239;


Ala, Ile, Met, or Thr for the amino acid at position 240;


Asp, Glu, Leu, Arg, Trp, or Tyr for the amino acid at position 241;


Leu, Glu, Leu, Gln, Arg, Trp, or Tyr for the amino acid at position 243;


His for the amino acid at position 244;


Ala for the amino acid at position 245;


Asp, Glu, His, or Tyr for the amino acid at position 246;


Ala, Phe, Gly, His, Ile, Leu, Met, Thr, Val, or Tyr for the amino acid at position 247;


Glu, His, Gln, or Tyr for the amino acid at position 249;


Glu or Gln for the amino acid at position 250;


Phe for the amino acid at position 251;


Phe, Met, or Tyr for the amino acid at position 254;


Glu, Leu, or Tyr for the amino acid at position 255;


Ala, Met, or Pro for the amino acid at position 256;


Asp, Glu, His, Ser, or Tyr for the amino acid at position 258;


Asp, Glu, His, or Tyr for the amino acid at position 260;


Ala, Glu, Phe, Ile, or Thr for the amino acid at position 262;


Ala, Ile, Met, or Thr for the amino acid at position 263;


Asp, Glu, Phe, Gly, His, Ile, Lys, Leu, Met, Asn, Pro, Gln, Arg, Ser, Thr, Trp, or Tyr for the amino acid at position 264;


Ala, Leu, Phe, Gly, His, Ile, Lys, Leu, Met, Asn, Pro, Gln, Arg, Ser, Thr, Val, Trp, or Tyr for the amino acid at position 265;


Ala, Ile, Met, or Thr for the amino acid at position 266;


Asp, Glu, Phe, His, Ile, Lys, Leu, Met, Asn, Pro, Gln, Arg, Thr, Val, Trp, or Tyr for the amino acid at position 267;


Asp, Glu, Phe, Gly, Ile, Lys, Leu, Met, Pro, Gln, Arg, Thr, Val, or Trp for the amino acid at position 268;


Phe, Gly, His, Ile, Lys, Leu, Met, Asn, Pro, Arg, Ser, Thr, Val, Trp, or Tyr for the amino acid at position 269;


Glu, Phe, Gly, His, Ile, Leu, Met, Pro, Gln, Arg, Ser, Thr, Trp, or Tyr for the amino acid at position 270;


Ala, Asp, Glu, Phe, Gly, His, Ile, Lys, Leu, Met, Asn, Gln, Arg, Ser, Thr, Val, Trp, or Tyr for the amino acid at position 271;


Asp, Phe, Gly, His, Ile, Lys, Leu, Met, Pro, Arg, Ser, Thr, Val, Trp, or Tyr for the amino acid at position 272;


Phe or Ile for the amino acid at position 273;


Asp, Glu, Phe, Gly, His, Ile, Leu, Met, Asn, Pro, Arg, Ser, Thr, Val, Trp, or Tyr for the amino acid at position 274;


Leu or Trp for the amino acid at position 275;


Asp, Glu, Phe, Gly, His, Ile, Leu, Met, Pro, Arg, Ser, Thr, Val, Trp, or Tyr for the amino acid at position 276;


Asp, Glu, Gly, His, Ile, Lys, Leu, Met, Asn, Pro, Gln, Arg, Ser, Thr, Val, or Trp for the amino acid at position 278;


Ala for the amino acid at position 279;


Ala, Gly, His, Lys, Leu, Pro, Gln, Trp, or Tyr for the amino acid at position 280;


Asp, Lys, Pro, or Tyr for the amino acid at position 281;


Glu, Gly, Lys, Pro, or Tyr for the amino acid at position 282;


Ala, Gly, His, Ile, Lys, Leu, Met, Pro, Arg, or Tyr for the amino acid at position 283;


Asp, Glu, Leu, Asn, Thr, or Tyr for the amino acid at position 284;


Asp, Glu, Lys, Gln, Trp, or Tyr for the amino acid at position 285;


Glu, Gly, Pro, or Tyr for the amino acid at position 286;


Asn, Asp, Glu, or Tyr for the amino acid at position 288;


Asp, Gly, His, Leu, Asn, Ser, Thr, Trp, or Tyr for the amino acid at position 290;


Asp, Glu, Gly, His, Ile, Gln, or Thr for the amino acid at position 291;


Ala, Asp, Glu, Pro, Thr, or Tyr for the amino acid at position 292;


Phe, Gly, His, Ile, Leu, Met, Asn, Pro, Arg, Ser, Thr, Val, Trp, or Tyr for the amino acid at position 293;


Phe, Gly, His, Ile, Lys, Leu, Met, Asn, Pro, Arg, Ser, Thr, Val, Trp, or Tyr for the amino acid at position 294;


Asp, Glu, Phe, Gly, His, Ile, Lys, Met, Asn, Pro, Arg, Ser, Thr, Val, Trp, or Tyr for the amino acid at position 295;


Ala, Asp, Glu, Gly, His, Ile, Lys, Leu, Met, Asn, Gln, Arg, Ser, Thr, or Val for the amino acid at position 296;


Asp, Glu, Phe, Gly, His, Ile, Lys, Leu, Met, Pro, Gln, Arg, Ser, Thr, Val, Trp, or Tyr for the amino acid at position 297;


Ala, Asp, Glu, Phe, His, Ile, Lys, Met, Asn, Gln, Arg, Thr, Val, Trp, or Tyr for the amino acid at position 298;


Ala, Asp, Glu, Phe, Gly, His, Ile, Lys, Leu, Met, Asn, Pro, Gln, Arg, Ser, Val, Trp, or Tyr for the amino acid at position 299;


Ala, Asp, Glu, Gly, His, Ile, Lys, Leu, Met, Asn, Pro, Gln, Arg, Ser, Thr, Val, or Trp for the amino acid at position 300;


Asp, Glu, His, or Tyr for the amino acid at position 301;


Ile for the amino acid at position 302;


Asp, Gly, or Tyr for the amino acid at position 303;


Asp, His, Leu, Asn, or Thr for the amino acid at position 304;


Glu, Ile, Thr, or Tyr for the amino acid at position 305;


Ala, Asp, Asn, Thr, Val, or Tyr for the amino acid at position 311;


Phe for the amino acid at position 313;


Leu for the amino acid at position 315;


Glu or Gln for the amino acid at position 317;


His, Leu, Asn, Pro, Gln, Arg, Thr, Val, or Tyr for the amino acid at position 318;


Asp, Phe, Gly, His, Ile, Leu, Asn, Pro, Ser, Thr, Val, Trp, or Tyr for the amino acid at position 320;


Ala, Asp, Phe, Gly, His, Ile, Pro, Ser, Thr, Val, Trp, or Tyr for the amino acid at position 322;


Ile for the amino acid at position 323;


Asp, Phe, Gly, His, Ile, Leu, Met, Pro, Arg, Thr, Val, Trp, or Tyr for the amino acid at position 324;


Ala, Asp, Glu, Phe, Gly, His, Ile, Lys, Leu, Met, Pro, Gln, Arg, Ser, Thr, Val, Trp, or Tyr for the amino acid at position 325;


Ala, Asp, Glu, Gly, Ile, Leu, Met, Asn, Pro, Gln, Ser, Thr, Val, Trp, or Tyr for the amino acid at position 326;


Ala, Asp, Glu, Phe, Gly, His, Ile, Lys, Leu, Met, Asn, Pro, Arg, Thr, Val, Trp, or Tyr for the amino acid at position 327;


Ala, Asp, Glu, Phe, Gly, His, Ile, Lys, Met, Asn, Pro, Gln, Arg, Ser, Thr, Val, Trp, or Tyr for the amino acid at position 328;


Asp, Glu, Phe, Gly, His, Ile, Lys, Leu, Met, Asn, Gln, Arg, Ser, Thr, Val, Trp, or Tyr for the amino acid at position 329;


Cys, Glu, Phe, Gly, His, Ile, Lys, Leu, Met, Asn, Pro, Arg, Ser, Thr, Val, Trp, or Tyr for the amino acid at position 330;


Asp, Phe, His, Ile, Leu, Met, Gln, Arg, Thr, Val, Trp, or Tyr for the amino acid at position 331;


Ala, Asp, Glu, Phe, Gly, His, Lys, Leu, Met, Asn, Pro, Gln, Arg, Ser, Thr, Val, Trp, or Tyr for the amino acid at position 332;


Ala, Asp, Glu, Phe, Gly, His, Ile, Leu, Met, Pro, Ser, Thr, Val, or Tyr for the amino acid at position 333;


Ala, Glu, Phe, Ile, Leu, Pro, or Thr for the amino acid at position 334;


Asp, Phe, Gly, His, Ile, Leu, Met, Asn, Pro, Arg, Ser, Val, Trp, or Tyr for the amino acid at position 335;


Glu, Lys, or Tyr for the amino acid at position 336;


Glu, His, or Asn for the amino acid at position 337;


Asp, Phe, Gly, Ile, Lys, Met, Asn, Gln, Arg, Ser, or Thr for the amino acid at position 339;


Ala or Val for the amino acid at position 376;


Gly or Lys for the amino acid at position 377;


Asp for the amino acid at position 378;


Asn for the amino acid at position 379;


Ala, Asn, or Ser for the amino acid at position 380;


Ala or Ile for the amino acid at position 382;


Glu for the amino acid at position 385;


Thr for the amino acid at position 392;


Leu for the amino acid at position 396;


Lys for the amino acid at position 421;


Asn for the amino acid at position 427;


Phe or Leu for the amino acid at position 428;


Met for the amino acid at position 429;


Trp for the amino acid at position 434;


Ile for the amino acid at position 436; and


Gly, His, Ile, Leu, or Tyr for the amino acid at position 440;


as indicated by EU numbering;


[22] the antigen-binding molecule of any one of [1] to [18], wherein the Fc region has a higher binding activity toward an inhibitory Fcγ receptor than toward an activating Fcγ receptor;


[23] the antigen-binding molecule of [22], wherein the inhibitory Fcγ receptor is human FcγRIIb;


[24] the antigen-binding molecule of [21] or [22], wherein the activating Fcγ receptor is human FcγRIa, human FcγRIIa (R), human FcγRIIa (H), human FcγRIIIa (V), or human FcγRIIIa (F);


[25] the antigen-binding molecule of any one of [22] to [24], wherein the amino acid at position 238 or 328 (EU numbering) in the Fc region is different from the amino acid in the native human IgG Fc region;


[26] the antigen-binding molecule of [25], wherein the amino acid at position 238 of the Fc region is Asp or the amino acid at position 328 of the Fc region is Glu as indicated by EU numbering;


[27] the antigen-binding molecule of [25] or [26], wherein the amino acid sequence of the Fc region comprises at least one or more amino acids selected from the group consisting of:


Asp for the amino acid at position 233;


Trp or Tyr for the amino acid at position 234;


Ala, Asp, Glu, Leu, Met, Phe, Trp, or Tyr for the amino acid at position 237;


Asp for the amino acid at position 239;


Ala, Gln, or Val for the amino acid at position 267;


Asn, Asp, or Glu for the amino acid at position 268;


Gly for the amino acid at position 271;


Ala, Asn, Asp, Gln, Glu, Leu, Met, Ser, or Thr for the amino acid at position 326;


Arg, Lys, or Met for the amino acid at position 330;


Ile, Leu, or Met for the amino acid at position 323; and


Asp for the amino acid at position 296;


as indicated by EU numbering;


[28] a pharmaceutical composition comprising the antigen-binding molecule of any one of [1] to


[27] as an active ingredient;


[29] use of the antigen-binding molecule of any one of [1] to [27] for eliminating an aggregated antigen from plasma;


[30] the use of the antigen-binding molecule of [29], wherein the aggregated antigen is eliminated in preference to an unaggregated antigen;


[31] a method of screening for an antigen-binding molecule which binds to an aggregated antigen and has a function of eliminating the aggregated antigen from plasma, which comprises step (a) below:

  • (a) selecting an antigen-binding molecule whose antigen-binding activity to an aggregated antigen under an intracellular ion concentration condition is lower than the binding activity under an extracellular ion concentration condition;


    [32] the screening method of [31], which further comprises the step(s) of:
  • (i) selecting an antigen-binding molecule whose binding activity to an aggregated antigen is higher than the binding activity to an unaggregated antigen under an extracellular ion concentration condition; and/or
  • (ii) selecting an antigen-binding molecule whose binding activity to an Fcγ receptor or an FcRn of a complex formed between an aggregated antigen and the antigen-binding molecule becomes higher than the binding activity to an Fcγ receptor or an FcRn of a complex formed between an unaggregated antigen and the antigen-binding molecule under an extracellular ion concentration condition;


    [33] a method for producing an antigen-binding molecule which binds to an aggregated antigen and has a function of eliminating the aggregated antigen from plasma, which comprises steps (a) to (c) below:
  • (a) selecting an antigen-binding molecule whose antigen-binding activity to an aggregated antigen under an intracellular ion concentration condition is lower than the binding activity under an extracellular ion concentration condition;
  • (b) culturing a host cell comprising a vector that carries a gene encoding the antigen-binding molecule selected in step (a) mentioned above; and
  • (c) isolating an antigen-binding molecule from the culture obtained in step (b) mentioned above; and


    [34] a method for producing an antigen-binding molecule which binds to an aggregated antigen and has a function of eliminating the aggregated antigen from plasma, which comprises steps (a) to (c) below:
  • (a) selecting an antigen-binding molecule whose antigen-binding activity to an aggregated antigen under an intracellular ion concentration condition is lower than the binding activity under an extracellular ion concentration condition;
  • (b) (i) selecting an antigen-binding molecule whose binding activity to an aggregated antigen is higher than the binding activity to an unaggregated antigen under an extracellular ion concentration condition, and/or (ii) selecting an antigen-binding molecule whose binding activity to an Fcγ receptor or an FcRn of a complex formed between an aggregated antigen and the antigen-binding molecule becomes higher than the binding activity to an Fcγ receptor or an FcRn of a complex formed between an unaggregated antigen and the antigen-binding molecule under an extracellular ion concentration condition;
  • (c) culturing a host cell comprising a vector that carries a gene encoding the antigen-binding molecule selected in steps (a) and (b) mentioned above; and
  • (d) isolating an antigen-binding molecule from the culture obtained in step (c) mentioned above.





BRIEF DESCRIPTION OF THE DRAWINGS


FIG. 1 is a diagram showing that an antibody with pH-dependent binding repeatedly binds to soluble antigens. (i) An antibody binds to soluble antigens; (ii) the antibody is non-specifically internalized into a cell by pinocytosis; (iii) the antibody binds to FcRn within the endosome, and the soluble antigens dissociate from the antibody; (iv) the soluble antigens are transferred to the lysosome and degraded; (v) after dissociation from the soluble antigens, the antibody is recycled to the plasma via FcRn; (vi) the recycled antibody can bind to soluble antigens again.



FIG. 2 is a diagram showing that enhancing FcRn binding under neutral conditions results in improving the effect of an antibody with pH-dependent binding to repeatedly bind to antigens: (i) an antibody binds to soluble antigens; (ii) the antibody is internalized into a cell by pinocytosis via FcRn; (iii) the soluble antigens dissociate from the antibody in the endosome; (iv) the soluble antigens are transferred into the lysosome and degraded; (v) after dissociation of the soluble antigens, the antibody is recycled to the plasma via FcRn; and (vi) the recycled antibody can bind to soluble antigens again.



FIG. 3 shows a result of SEC analysis performed on aggregated human IgA.



FIG. 4 presents Biacore sensorgrams showing the interaction of anti-human IgA antibodies with monomeric human IgA and with aggregated human IgA.



FIG. 5 shows a time course of the plasma antibody concentration in normal mice of the GA2−IgG1 antibody-administered group.



FIG. 6 shows a time course of the plasma monomeric human IgA concentration in normal mice of the group administered only with monomeric human IgA, and in normal mice of the monomeric human IgA+GA2-IgG1 antibody-administered group.



FIG. 7 shows a time course of the aggregated human IgA concentration in plasma of normal mice in the group administered only with aggregated human IgA, and in the aggregated human IgA+GA2−IgG1 antibody-administered group.



FIG. 8 shows a time course of “human IgA concentration when human IgA+GA2−IgG1 is administration/human IgA concentration when only human IgA is administered”



FIG. 9 is a diagram showing an example that an aggregated antigen is efficiently incorporated into the cell by binding to receptors multivalently and strongly through formation of a large immune complex by polyvalent binding of multiple antibodies to the aggregated antigen.



FIG. 10 is a diagram showing an example that incorporation of unaggregated antigens into the cell is not efficient because the unaggregated antigens do not form a large immune complex and their binding to the receptor is weak.





MODE FOR CARRYING OUT THE INVENTION

The definitions and detailed description below are provided to help the understanding of the present invention illustrated herein.


Aggregated Antigen

In the present invention, aggregated antigen refers to a molecule in a state at which two or more of a molecule (monomer) present in a normal biological fluid have become aggregated or multimerized. An aggregated antigen may be a molecule in which monomers having the same three-dimensional structure (protein secondary structure or tertiary structure) are aggregated or multimerized as compared to antigens normally present in in biological fluid, or it may be a molecule in which partially or totally degenerated molecules as compared to the monomer are aggregated or multimerized. Furthermore, an aggregated antigen may be a molecule in which a mixture of two is aggregated or multimerized. In the aggregated antigens, another type of antigen that does not bind to the antigen-binding molecule may also be present. A target antigen of an antigen-binding molecule of the present invention is not particularly limited as long as the antigen aggregates, but an antigen that aggregates in a pathological condition is preferred, and an antigen whose aggregated form is a disease-causing substance is more preferred. Examples of such antigens include the antigens shown later in Table 7. Herein, the antigen that has aggregated may be referred to as an aggregated antigen or a multimeric antigen.


Biological fluid in the present invention refers to all fluids that fill the space between the vasculature or tissues/cells in an organism where an aggregated antigen is present in its pathological condition. Specific examples include plasma, interstitial fluid, cerebrospinal fluid, spinal fluid, puncture liquid, synovial fluid, alveolar fluid (bronchoalveolar lavage fluid), lymph, ascites, pleural fluid, pericardial fluid, cyst fluid, aqueous humor (hydatoid), and such.


Methods of preparing such aggregated antigens include the methods below: (1) purifying an aggregated antigen by chromatography or such from plasma containing the aggregated antigen; (2) chemically crosslinking a monomeric antigen by chemical crosslinking agents such as SPDP (N-Succinimidyl 3-(2-pyridyldithio)propionate), and purifying the aggregated antigen by chromatography or such; and (3) purifying by chromatography or such an aggregated antigen formed through overall or partial chemical degeneration of a monomeric antigen by thermal treatment or acid treatment.


Amino Acids

Herein, amino acids are described in one- or three-letter codes or both, for example, Ala/A, Leu/L, Arg/R, Lys/K, Asn/N, Met/M, Asp/D, Phe/F, Cys/C, Pro/P, Gln/Q, Ser/S, Glu/E, Thr/T, Gly/G, Trp/W, His/H, Tyr/Y, Ile/I, or Val/V.


Alteration of Amino Acids

For amino acid alteration in the amino acid sequence of an antigen-binding molecule, known methods such as site-directed mutagenesis methods (Kunkel et al. (Proc. Natl. Acad. Sci. USA (1985) 82: 488-492)) and overlap extension PCR may be appropriately adopted. Furthermore, several known methods may also be adopted as amino acid alteration methods for substitution to non-natural amino acids (Annu Rev. Biophys. Biomol. Struct. (2006) 35: 225-249; and Proc. Natl. Acad. Sci. U.S.A. (2003) 100(11): 6353-6357). For example, it is suitable to use a cell-free translation system (Clover Direct (Protein Express)) containing a tRNA that has a non-natural amino acid bound to a complementary amber suppressor tRNA of the UAG codon (amber codon) which is one of the stop codons.


Herein, the meaning of the term “and/or” when describing the site of amino acid alteration includes every combination where “and” and “or” are suitably combined. Specifically, for example, “the amino acids at positions 33, 55, and/or 96 are substituted” includes the following variation of amino acid alterations:


amino acid(s) at (a) position 33; (b) position 55; (c) position 96; (d) positions 33 and 55; (e) positions 33 and 96; (f) positions 55 and 96; and (g) positions 33, 55, and 96.


Epitopes

“Epitope” means an antigenic determinant in an antigen, and refers to an antigen site to which the antigen-binding domain of an antigen-binding molecule disclosed herein binds. Thus, for example, the epitope can be defined according to its structure. Alternatively, the epitope may be defined according to the antigen-binding activity of an antigen-binding molecule that recognizes the epitope. When the antigen is a peptide or polypeptide, the epitope can be specified by the amino acid residues forming the epitope. Alternatively, when the epitope is a sugar chain, the epitope can be specified by its specific sugar chain structure.


A linear epitope is an epitope that contains an epitope whose primary amino acid sequence has been recognized. Such a linear epitope typically contains at least three and most commonly at least five, for example, about 8 to about 10 or 6 to 20 amino acids in a specific sequence.


In contrast to the linear epitope, a “conformational epitope” is an epitope in which the primary amino acid sequence containing the epitope is not the only determinant of the recognized epitope (for example, the primary amino acid sequence of a conformational epitope is not necessarily recognized by an epitope-defining antibody). Conformational epitopes may contain a greater number of amino acids compared to linear epitopes. A conformational epitope-recognizing antibody recognizes the three-dimensional structure of a peptide or protein. For example, when a protein molecule folds and forms a three-dimensional structure, amino acids and/or polypeptide main chains that form a conformational epitope become aligned, and the epitope is made recognizable by the antibody. Methods for determining epitope conformations include, for example, X ray crystallography, two-dimensional nuclear magnetic resonance, site-specific spin labeling, and electron paramagnetic resonance, but are not limited thereto. See, for example, Epitope Mapping Protocols in Methods in Molecular Biology (1996), Vol. 66, Morris (ed.).


The structure of the antigen-binding domain which binds to an epitope is called a paratope. An epitope and a paratope bind with stability through the action of hydrogen bonds, electrostatic force, van der Waals force, hydrophobic bonds, and such between the epitope and the paratope. This strength of binding between the epitope and paratope is called affinity. The total sum of binding strength when a plurality of epitopes and a plurality of paratopes bind is referred to as avidity. When an antibody comprising a plurality of paratopes (i.e., multivalent antibody) or such binds to a plurality of epitopes, the affinity acts additively or synergistically, and therefore avidity becomes higher than affinity.


Binding Activity

Examples of a method for assessing epitope binding by a test antigen-binding molecule containing an antigen-binding domain directed to an antigen are described below; but a method is not limited thereto.


For example, whether a test antigen-binding molecule containing an antigen-binding domain against an antigen recognizes a linear epitope in the antigen molecule can be confirmed for example as mentioned below. For example, a linear peptide comprising an amino acid sequence forming the antigen is synthesized for the above purpose. The peptide can be synthesized chemically, or obtained by genetic engineering techniques using a cDNA encoding the antigen. Then, a test antigen-binding molecule containing an antigen-binding domain toward the antigen is assessed for its binding activity towards the linear peptide. For example, an ELISA using an immobilized linear peptide as an antigen can be performed to evaluate the binding activity of the antigen-binding molecule towards the peptide. Alternatively, the binding activity towards a linear peptide can be assessed based on the level of inhibition by the linear peptide of the binding of the antigen-binding molecule toward cells expressing on its surface the antigen. These tests can demonstrate the binding activity of the antigen-binding molecule towards the linear peptide.


Recognition of a conformational epitope by a test antigen-binding molecule comprising an antigen-binding domain targeting an antigen may be confirmed as stated below. For the above-mentioned objective, as described herein, a general genetic recombination technique is used to transfer an antigen-encoding recombinant gene into host cells (for example, animal cells, insect cells, or yeast cells) that enable formation of the native conformational epitope in the antigen. Antigen containing the conformational epitope is prepared from the culture of recombinant cells produced in this manner. Recognition of a conformational epitope by a test antigen-binding molecule comprising an antigen-binding domain targeting antigen is, for example, when the test antigen-binding molecule binds strongly to the antigen when it is contacted with immobilized antigen containing the conformational epitope, while the antigen-binding molecule does not substantively bind to a linear peptide comprising an amino acid sequence constituting the amino acid sequence of the immobilized antigen. Alternatively, it is also possible to use, instead of the above-mentioned linear peptide, the test IgA-targeting antigen-binding molecule that has been denatured by a reducing agent that cleaves disulfide bonds, such as dithiothreitol, dithioerythritol, β-mercaptoethanol, phosphines, and sodium borohydride, and/or chaotropic agents such as surfactants including guanidine hydrochloride, urea, and sodium lauryl sulfate. Here, the phrase “does not substantively bind” refers to a binding activity not greater than 80%, normally not greater than 50%, preferably not greater than 30%, or particularly preferably not greater than 15% of the human-IgA-binding activity.


Methods for assaying the binding activity toward an antigen of a test antigen-binding molecule containing an antigen-binding domain against the antigen include, for example, the methods described in Antibodies: A Laboratory Manual (Ed Harlow, David Lane, Cold Spring Harbor Laboratory (1988) 359-420). Specifically, the assessment can be performed based on the principle of ELISA or EIA using IgA as antigen.


In the ELISA format, the binding activity of a test antigen-binding molecule containing an antigen-binding domain towards the antigen can be assessed quantitatively by comparing the levels of signal generated by enzymatic reaction. Specifically, a test antigen-binding molecule is added to an ELISA plate onto which an antigen has been immobilized. Then, the test antigen-binding molecule that bound to an antigen immobilized on the plate is detected using an enzyme-labeled antibody that recognizes the test antigen-binding molecule. In the ELISA, a serial dilution of the test antigen-binding molecule can be prepared and the antibody binding titer toward an antigen is determined to compare the binding activity of the test antigen-binding molecule towards the antigen.


The binding of a test antigen-binding molecule towards an antigen expressed on the surface of cells suspended in buffer or the like can be detected using a flow cytometer. Known flow cytometers include, for example, the following devices:


FACSCanto™ II
FAC SAria™
FAC SArray™
FACSVantage™ SE

FACSCalibur™ (all are trade names of BD Biosciences)


EPICS ALTRA HyPerSort
Cytomics FC 500
EPICS XL-MCL ADC EPICS XL ADC

Cell Lab Quanta/Cell Lab Quanta SC (all are trade names of Beckman Coulter).


Alternatively, preferable methods for assaying the binding activity towards an antigen of a test antigen-binding molecule containing an antigen-binding domain against an antigen include, for example, the following method. First, antigen-expressing cells are reacted with a test antigen-binding molecule, and then this is stained with an FITC-labeled secondary antibody that recognizes the antigen-binding molecule. The test antigen-binding molecule is appropriately diluted with a suitable buffer to prepare the molecule at a desired concentration. For example, the molecule can be used at a concentration within the range of 10 μg/ml to 10 ng/ml. Then, the fluorescence intensity and cell count are determined using FACSCalibur (BD). The fluorescence intensity obtained by analysis using the CELL QUEST Software (BD), i.e., the Geometric Mean value, reflects the quantity of antibody bound to cells. That is, the binding activity of a test antigen-binding molecule, which is represented by the quantity of the test antigen-binding molecule bound, can be determined by measuring the Geometric Mean value.


Whether a test antigen-binding molecule containing a binding domain towards an antigen shares a common epitope with another antigen-binding molecule can be assessed based on the competition between the two molecules for the same epitope. The competition between antigen-binding molecules can be detected by cross-blocking assay or the like. For example, the competitive ELISA assay is a preferred cross-blocking assay.


Specifically, in cross-blocking assay, the antigen immobilized to the wells of a microtiter plate is pre-incubated in the presence or absence of a candidate competitor antigen-binding molecule, and then a test antigen-binding molecule is added thereto. The quantity of test antigen-binding molecule bound to the antigen in the wells is indirectly correlated with the binding ability of a candidate competitor antigen-binding molecule that competes for the binding to the same epitope. That is, the greater the affinity of the competitor antigen-binding molecule for the same epitope, the lower the binding activity of the test antigen-binding molecule towards the antigen-coated wells.


The quantity of the test antigen-binding molecule bound to the wells via the antigen can be readily determined by labeling the antigen-binding molecule in advance. For example, a biotin-labeled antigen-binding molecule is measured using an avidin/peroxidase conjugate and appropriate substrate. In particular, cross-blocking assay that uses enzyme labels such as peroxidase is called “competitive ELISA assay”. The antigen-binding molecule can also be labeled with other labeling substances that enable detection or measurement. Specifically, radiolabels, fluorescent labels, and such are known.


When the candidate competitor antigen-binding molecule can block the antigen binding by a test antigen-binding molecule by at least 20%, preferably at least 20 to 50%, and more preferably at least 50% compared to the binding activity in a control experiment conducted in the absence of the candidate competitor antigen-binding molecule, the test antigen-binding molecule is determined to substantially bind to the same epitope bound by the competitor antigen-binding molecule, or compete for the binding to the same epitope.


When the structure of an epitope bound by a test antigen-binding molecule containing a binding domain towards an antigen has already been identified, whether the test and control antigen-binding molecules share a common epitope can be assessed by comparing the binding activities of the two antigen-binding molecules towards a peptide prepared by introducing amino acid mutations into the peptide forming the epitope.


To measure the above binding activities, for example, the binding activities of test and control antigen-binding molecules towards a linear peptide into which a mutation is introduced are compared in the above ELISA format. Besides the ELISA methods, the binding activity towards the mutant peptide bound to a column can be determined by flowing test and control antigen-binding molecules in the column, and then quantifying the antigen-binding molecule eluted in the elution solution. Methods for adsorbing a mutant peptide to a column, for example, in the form of a GST fusion peptide, are known.


Alternatively, when the identified epitope in an antigen-binding molecule expressed on a cell is a conformational epitope, whether test and control antigen-binding molecules share a common epitope can be assessed, for example, by the following method. First, cells expressing an antigen of interest and cells expressing an antigen with a mutation introduced into the epitope are prepared. The test and control antigen-binding molecules are added to a cell suspension prepared by suspending these cells in an appropriate buffer such as PBS. Then, the cell suspensions are appropriately washed with a buffer, and an FITC-labeled antibody that recognizes the test and control antigen-binding molecules is added thereto. The fluorescence intensity and number of cells stained with the labeled antibody are determined using FACSCalibur (BD). The test and control antigen-binding molecules are appropriately diluted using a suitable buffer, and used at desired concentrations. For example, they may be used at a concentration within the range of 10 μg/ml to 10 ng/ml. The fluorescence intensity determined by analysis using the CELL QUEST Software (BD), i.e., the Geometric Mean value, reflects the quantity of labeled antibody bound to cells. That is, the binding activities of the test and control antigen-binding molecules, which are represented by the quantity of labeled antibody bound, can be determined by measuring the Geometric Mean value.


In the present invention, whether the binding activity to an aggregated antigen is higher than the binding activity to an unaggregated antigen can be confirmed by comparing the binding activity to the aggregated antigen and the binding activity to the unaggregated antigen according to the above-described method.


Antigen-Binding Domain

Herein, an “antigen-binding domain” may be of any structure as long as it binds to an antigen of interest. Such domains preferably include, for example:


antibody heavy-chain and light-chain variable regions;


a module of about 35 amino acids called A domain which is contained in the in vivo cell membrane protein Avimer (International Publication No. WO 2004/044011, International Publication No. WO 2005/040229);


Adnectin containing the 1 OFn3 domain which binds to the protein moiety of fibronectin, a glycoprotein expressed on cell membrane (International Publication No. WO 2002/032925);


Affibody which is composed of a 58-amino acid three-helix bundle based on the scaffold of the IgG-binding domain of Protein A (International Publication No. WO 1995/001937);


Designed Ankyrin Repeat proteins (DARPins) which are a region exposed on the molecular surface of ankyrin repeats (AR) having a structure in which a subunit consisting of a turn comprising 33 amino acid residues, two antiparallel helices, and a loop is repeatedly stacked (International Publication No. WO 2002/020565);


Anticalins and such, which are domains consisting of four loops that support one side of a barrel structure composed of eight circularly arranged antiparallel strands that are highly conserved among lipocalin molecules such as neutrophil gelatinase-associated lipocalin (NGAL) (International Publication No. WO 2003/029462); and


the concave region formed by the parallel-sheet structure inside the horseshoe-shaped structure constituted by stacked repeats of the leucine-rich-repeat (LRR) module of the variable lymphocyte receptor (VLR) which does not have the immunoglobulin structure and is used in the system of acquired immunity in jawless vertebrate such as lampery and hagfish (International Publication No. WO 2008/016854). Preferred antigen-binding domains of the present invention include, for example, those having antibody heavy-chain and light-chain variable regions. Preferred examples of antigen-binding domains include “single chain Fv (scFv)”, “single chain antibody”, “Fv”, “single chain Fv 2 (scFv2)”, “Fab”, and “F(ab′)2”.


Immune Complex

Immune complex refers to a structure produced when at least one unaggregated antigen or aggregated antigen and at least one antigen-binding molecule bind with each other to form a larger molecular-weight complex consisting of antigen(s) and antigen-binding molecule(s).


Antibody

Herein, “antibody” refers to a natural immunoglobulin or an immunoglobulin produced by partial or complete synthesis. Antibodies can be isolated from natural sources such as naturally-occurring plasma and serum, or culture supernatants of antibody-producing hybridomas. Alternatively, antibodies can be partially or completely synthesized using techniques such as genetic recombination. Preferred antibodies include, for example, antibodies of an immunoglobulin isotype or subclass belonging thereto. Known human immunoglobulins include antibodies of the following nine classes (isotypes): IgG1, IgG2, IgG3, IgG4, IgA1, IgA2, IgD, IgE, and IgM. Of these isotypes, antibodies of the present invention include IgG1, IgG2, IgG3, and IgG4. A number of allotype sequences of human IgG1, human IgG2, human IgG3, and human IgG4 constant regions due to gene polymorphisms are described in “Sequences of proteins of immunological interest”, NIH Publication No. 91-3242. Any of such sequences may be used in the present invention. In particular, for the human IgG1 sequence, the amino acid sequence at positions 356 to 358 as indicated by EU numbering may be DEL or EEM. Several allotype sequences due to genetic polymorphisms have been described in “Sequences of proteins of immunological interest”, NIH Publication No. 91-3242 for the human Igκ (Kappa) constant region and human Igλ, (Lambda) constant region, and any of the sequences may be used in the present invention.


Methods for producing an antibody with desired binding activity are known to those skilled in the art.


Antibodies can be obtained as polyclonal or monoclonal antibodies using known methods. Mammal-derived monoclonal antibodies include antibodies produced by hybridomas or host cells transformed with an expression vector carrying an antibody gene by genetic engineering techniques. Meanwhile, “humanized antibodies” or “chimeric antibodies” are included in the monoclonal antibodies of the present invention.


Monoclonal antibody-producing hybridomas can be produced using known techniques, for example, as described below. Specifically, mammals are immunized by conventional immunization methods using a sensitizing antigen. Resulting immune cells are fused with known parental cells by conventional cell fusion methods. Then, hybridomas producing an antibody of interest can be selected by screening for monoclonal antibody-producing cells using conventional screening methods. There is no particular limitation on the mammals to be immunized with the sensitizing antigen. However, it is preferable to select the mammals by considering their compatibility with the parent cells to be used for cell fusion. In general, rodents such as mice, rats, and hamsters, rabbits, and monkeys are preferably used.


The above animals are immunized with a sensitizing antigen by known methods. Generally performed immunization methods include, for example, intraperitoneal or subcutaneous injection of a sensitizing antigen into mammals. Specifically, a sensitizing antigen is appropriately diluted with PBS (Phosphate-Buffered Saline), physiological saline, or the like. If desired, a conventional adjuvant such as Freund's complete adjuvant is mixed with the antigen, and the mixture is emulsified. Then, the sensitizing antigen is administered to a mammal several times at 4- to 21-day intervals. Appropriate carriers may be used in immunization with the sensitizing antigen. In particular, when a low-molecular-weight partial peptide is used as the sensitizing antigen, it is sometimes desirable to couple the sensitizing antigen peptide to a carrier protein such as albumin or keyhole limpet hemocyanin for immunization.


Alternatively, hybridomas producing a desired antibody can be prepared using DNA immunization as mentioned below. DNA immunization is an immunization method that confers immunostimulation by expressing a sensitizing antigen in an animal immunized as a result of administering a vector DNA constructed to allow expression of an antigen protein-encoding gene in the animal. As compared to conventional immunization methods in which a protein antigen is administered to animals to be immunized, DNA immunization is expected to be superior in that:


in the case the antigen is a membrane protein, immunostimulation can be provided while retaining the structure of the membrane protein; and


there is no need to purify the antigen for immunization.


In order to prepare a monoclonal antibody of the present invention using DNA immunization, first, a DNA expressing an antigen protein is administered to an animal to be immunized. The antigen protein-encoding DNA can be synthesized by known methods such as PCR. The obtained DNA is inserted into an appropriate expression vector, and then this is administered to an animal to be immunized. Preferably used expression vectors include, for example, commercially-available expression vectors such as pcDNA3.1. Vectors can be administered to an organism using conventional methods. For example, DNA immunization is performed by using a gene gun to introduce expression vector-coated gold particles into cells in the body of an animal to be immunized.


After immunizing a mammal as described above, an increase in the titer of an antibody against a desired antigen is confirmed in the serum. Then, immune cells are collected from the mammal, and then subjected to cell fusion. In particular, splenocytes are preferably used as immune cells.


A mammalian myeloma cell is used as a cell to be fused with the above-mentioned immune cells. The myeloma cells preferably comprise a suitable selection marker for screening. A selection marker confers characteristics to cells for their survival (or death) under a specific culture condition. Hypoxanthine-guanine-phosphoribosyltransferase deficiency (hereinafter abbreviated as HGPRT deficiency) and thymidine kinase deficiency (hereinafter abbreviated as TK deficiency) are known as selection markers. Cells with HGPRT or TK deficiency have hypoxanthine-aminopterin-thymidine sensitivity (hereinafter abbreviated as HAT sensitivity). HAT-sensitive cells cannot synthesize DNA in a HAT selection medium, and are thus killed. However, when the cells are fused with normal cells, they can continue DNA synthesis using the salvage pathway of the normal cells, and therefore they can grow even in the HAT selection medium.


HGPRT-deficient and TK-deficient cells can be selected in a medium containing 6-thioguanine, 8-azaguanine (hereinafter abbreviated as 8AG), or 5′-bromodeoxyuridine, respectively. Normal cells are killed because they incorporate these pyrimidine analogs into their DNA. Meanwhile, cells that are deficient in these enzymes can survive in the selection medium, since they cannot incorporate these pyrimidine analogs. In addition, a selection marker referred to as G418 resistance provided by the neomycin-resistant gene confers resistance to 2-deoxystreptamine antibiotics (gentamycin analogs). Various types of myeloma cells that are suitable for cell fusion are known.


For example, myeloma cells including the following cells can be preferably used:


P3(P3×63Ag8.653) (J. Immunol. (1979) 123 (4), 1548-1550);
P3×63Ag8U.1 (Current Topics in Microbiology and Immunology (1978)81, 1-7);

NS-1 (C. Eur. J. Immunol. (1976)6 (7), 511-519);


MPC-11 (Cell (1976) 8 (3), 405-415);
SP2/0 (Nature (1978) 276 (5685), 269-270);

FO (J. Immunol. Methods (1980) 35 (1-2), 1-21);


S194/5.XXO.BU.1 (J. Exp. Med. (1978) 148 (1), 313-323);


R210 (Nature (1979) 277 (5692), 131-133), etc.

Cell fusions between the immunocytes and myeloma cells are essentially carried out using known methods, for example, a method by Kohler and Milstein et al. (Methods Enzymol. (1981) 73: 3-46).


More specifically, cell fusion can be carried out, for example, in a conventional culture medium in the presence of a cell fusion-promoting agent. The fusion-promoting agents include, for example, polyethylene glycol (PEG) and Sendai virus (HVJ). If required, an auxiliary substance such as dimethyl sulfoxide is also added to improve fusion efficiency.


The ratio of immune cells to myeloma cells may be determined at one's own discretion, preferably, for example, one myeloma cell for every one to ten immunocytes. Culture media to be used for cell fusions include, for example, media that are suitable for the growth of myeloma cell lines, such as RPMI1640 medium and MEM medium, and other conventional culture medium used for this type of cell culture. In addition, serum supplements such as fetal calf serum (FCS) may be preferably added to the culture medium.


For cell fusion, predetermined amounts of the above immune cells and myeloma cells are mixed well in the above culture medium. Then, a PEG solution (for example, the average molecular weight is about 1000 to 6000) prewarmed to about 37° C. is added thereto at a concentration of generally 30% to 60% (w/v). This is gently mixed to produce desired fusion cells (hybridomas). Then, an appropriate culture medium mentioned above is gradually added to the cells, and this is repeatedly centrifuged to remove the supernatant. Thus, cell fusion agents and such which are unfavorable to hybridoma growth can be removed.


The hybridomas thus obtained can be selected by culture using a conventional selective medium, for example, HAT medium (a culture medium containing hypoxanthine, aminopterin, and thymidine). Cells other than the desired hybridomas (non-fused cells) can be killed by continuing culture in the above HAT medium for a sufficient period of time (typically, the period is several days to several weeks). Then, hybridomas producing the desired antibody are screened and singly cloned by conventional limiting dilution methods.


The hybridomas thus obtained can be selected using a selection medium based on the selection marker possessed by the myeloma used for cell fusion. For example, HGPRT- or TK-deficient cells can be selected by culture using the HAT medium (a culture medium containing hypoxanthine, aminopterin, and thymidine). Specifically, when HAT-sensitive myeloma cells are used for cell fusion, cells successfully fused with normal cells can selectively proliferate in the HAT medium. Cells other than the desired hybridomas (non-fused cells) can be killed by continuing culture in the above HAT medium for a sufficient period of time. Specifically, desired hybridomas can be selected by culture for generally several days to several weeks. Then, hybridomas producing the desired antibody are screened and singly cloned by conventional limiting dilution methods.


Desired antibodies can be preferably selected and singly cloned by screening methods based on known antigen/antibody reaction. For example, the activity of an antibody to bind to immobilized antigen can be assessed based on the principle of ELISA. For example, antigen is immobilized to the wells of an ELISA plate. Culture supernatants of hybridomas are contacted with the IgA in the wells, and antibodies that bind to the IgA are detected. When the monoclonal antibodies are derived from mouse, antibodies bound to the cells can be detected using an anti-mouse immunoglobulin antibody. Hybridomas producing a desired antibody having the antigen-binding ability are selected by the above screening, and they can be cloned by a limiting dilution method or the like.


Monoclonal antibody-producing hybridomas thus prepared can be passaged in a conventional culture medium, and stored in liquid nitrogen for a long period.


The above hybridomas are cultured by a conventional method, and desired monoclonal antibodies can be prepared from the culture supernatants. Alternatively, the hybridomas are administered to and grown in compatible mammals, and monoclonal antibodies are prepared from the ascites. The former method is suitable for preparing antibodies with high purity.


Antibodies encoded by antibody genes that are cloned from antibody-producing cells such as the above hybridomas can also be preferably used. A cloned antibody gene is inserted into an appropriate vector, and this is introduced into a host to express the antibody encoded by the gene. Methods for isolating antibody genes, inserting the genes into vectors, and transforming host cells have already been established, for example, by Vandamme et al. (Eur. J. Biochem. (1990) 192(3), 767-775). Methods for producing recombinant antibodies are also known as described below.


For example, a cDNA encoding the variable region (V region) of an antibody of interest is prepared from hybridoma cells producing the antibody. For this purpose, total RNA is first extracted from hybridomas. Methods used for extracting mRNAs from cells include, for example:


the guanidine ultracentrifugation method (Biochemistry (1979) 18(24), 5294-5299), and


the AGPC method (Anal. Biochem. (1987) 162(1), 156-159)


Extracted mRNAs can be purified using the mRNA Purification Kit (GE Healthcare Bioscience) or such. Alternatively, kits for extracting total mRNA directly from cells, such as the QuickPrep mRNA Purification Kit (GE Healthcare Bioscience), are also commercially available. mRNAs can be prepared from hybridomas using such kits. cDNAs encoding the antibody V region can be synthesized from the prepared mRNAs using a reverse transcriptase. cDNAs can be synthesized using the AMV Reverse Transcriptase First-strand cDNA Synthesis Kit (Seikagaku Co.) or such. Furthermore, the SMART RACE cDNA amplification kit (Clontech) and the PCR-based 5′-RACE method (Proc. Natl. Acad. Sci. USA (1988) 85(23), 8998-9002; Nucleic Acids Res. (1989) 17(8), 2919-2932) can be appropriately used to synthesize and amplify cDNAs. In such a cDNA synthesis process, appropriate restriction enzyme sites described below may be introduced into both ends of a cDNA.


The cDNA fragment of interest is purified from the resulting PCR product, and then this is ligated to a vector DNA. A recombinant vector is thus constructed, and introduced into E. coli or such. After colony selection, the desired recombinant vector can be prepared from the colony-forming E. coll. Then, whether the recombinant vector has the cDNA nucleotide sequence of interest is tested by a known method such as the dideoxy nucleotide chain termination method.


The 5′-RACE method which uses primers to amplify the variable region gene is conveniently used for isolating the gene encoding the variable region. First, a 5′-RACE cDNA library is constructed by cDNA synthesis using RNAs extracted from hybridoma cells as a template. A commercially available kit such as the SMART RACE cDNA amplification kit is appropriately used to synthesize the 5′-RACE cDNA library.


The antibody gene is amplified by PCR using the prepared 5′-RACE cDNA library as a template. Primers for amplifying the mouse antibody gene can be designed based on known antibody gene sequences. The nucleotide sequences of the primers vary depending on the immunoglobulin subclass. Therefore, it is preferable that the subclass is determined in advance using a commercially available kit such as the Iso Strip mouse monoclonal antibody isotyping kit (Roche Diagnostics).


Specifically, for example, primers that allow amplification of genes encoding γ1, γ2a, γ2b, and γ3 heavy chains and κ and λ, light chains are used to isolate mouse IgG-encoding genes. In general, a primer that anneals to a constant region site close to the variable region is used as a 3′-side primer to amplify an IgG variable region gene. Meanwhile, a primer attached to a 5′ RACE cDNA library construction kit is used as a 5′-side primer.


PCR products thus amplified are used to reshape immunoglobulins composed of a combination of heavy and light chains. A desired antibody can be selected using the antigen-binding activity of a reshaped immunoglobulin as an indicator. For example, when the objective is to isolate an antibody against a desired antigen, it is more preferred that the binding of the antibody to antigen is specific. An antibody that binds to the antigen can be screened, for example, by the following steps:

  • (1) contacting a desired antigen with an antibody comprising the V region encoded by a cDNA isolated from a hybridoma;
  • (2) detecting the binding of the antibody to the antigen; and
  • (3) selecting an antibody that binds to the antigen-expressing cell.


Methods for detecting the binding of an antibody to antigen are known. Specifically, the binding of an antibody to antigen-expressing cells can be detected by the above-described techniques such as ELISA.


Preferred antibody screening methods that use the binding activity as an indicator also include panning methods using phage vectors. Screening methods using phage vectors are advantageous when the antibody genes are isolated from heavy-chain and light-chain subclass libraries from a polyclonal antibody-expressing cell population. Genes encoding the heavy-chain and light-chain variable regions can be linked by an appropriate linker sequence to form a single-chain Fv (scFv). Phages presenting scFv on their surface can be produced by inserting a gene encoding scFv into a phage vector. The phages are contacted with an antigen of interest. Then, a DNA encoding scFv having the binding activity of interest can be isolated by collecting phages bound to the antigen. This process can be repeated as necessary to enrich scFv having the binding activity of interest.


After isolation of the cDNA encoding the V region of the antibody of interest, the cDNA is digested with restriction enzymes that recognize the restriction sites introduced into both ends of the cDNA. Preferred restriction enzymes recognize and cleave a nucleotide sequence that occurs in the nucleotide sequence of the antibody gene at a low frequency. Furthermore, a restriction site for an enzyme that produces a sticky end is preferably introduced into a vector to insert a single-copy digested fragment in the correct orientation. The cDNA encoding the V region of the antibody is digested as described above, and this is inserted into an appropriate expression vector to construct an antibody expression vector. In this case, if a gene encoding the antibody constant region (C region) and a gene encoding the above V region are fused in-frame, a chimeric antibody is obtained. Herein, “chimeric antibody” means that the origin of the constant region is different from that of the variable region. Thus, in addition to mouse-human heterochimeric antibodies, human-human allochimeric antibodies are included in the chimeric antibodies of the present invention. A chimeric antibody expression vector can be constructed by inserting the above V region gene into an expression vector that already has the constant region. Specifically, for example, a recognition sequence for a restriction enzyme that excises the above V region gene can be appropriately placed on the 5′ side of an expression vector carrying a DNA encoding a desired antibody constant region (C region). A chimeric antibody expression vector is constructed by fusing in frame the two genes digested with the same combination of restriction enzymes.


To produce a monoclonal antibody, antibody genes are inserted into an expression vector so that the genes are expressed under the control of an expression regulatory region. The expression regulatory region for antibody expression includes, for example, enhancers and promoters. Furthermore, an appropriate signal sequence may be attached to the amino terminus so that the expressed antibody is secreted to the outside of cells. The expressed polypeptide is cleaved at the carboxyl terminus of the above sequence, and the resulting polypeptide is secreted to the outside of cells as a mature polypeptide. Then, appropriate host cells are transformed with the expression vector, and recombinant cells expressing the DNA encoding a desired antibody are obtained.


DNAs encoding the antibody heavy chain (H chain) and light chain (L chain) are separately inserted into different expression vectors to express the antibody gene. An antibody molecule having the H and L chains can be expressed by co-transfecting the same host cell with vectors into which the H-chain and L-chain genes are respectively inserted. Alternatively, host cells can be transformed with a single expression vector into which DNAs encoding the H and L chains are inserted (see International Publication No. WO 1994/011523).


There are various known host cell/expression vector combinations for antibody preparation by introducing isolated antibody genes into appropriate hosts. All of these expression systems are applicable to isolation of the antigen-binding molecules of the present invention. Appropriate eukaryotic cells used as host cells include animal cells, plant cells, and fungal cells. Specifically, the animal cells include, for example, the following cells.

  • (1) Mammalian cells: CHO (Chinese hamster ovary cell line), COS (Monkey kidney cell line), myeloma (Sp2/0, NSO, etc.), BHK (baby hamster kidney cell line), HEK293 (human embryonic kidney cell line with sheared adenovirus (Ad)5 DNA), PER.C6 cell (human embryonic retinal cell line transformed with the Adenovirus Type 5 (Ad5) ElA and ElB genes), Hela, Vero, or such (Current Protocols in Protein Science (May, 2001, Unit 5.9, Table 5.9.1)).
  • (2) Amphibian cells: Xenopus oocytes, or such.
  • (3) Insect cells: sf9, sf21, Tn5, or such.


In addition, as a plant cell, an antibody gene expression system using cells derived from the Nicotiana genus such as Nicotiana tabacum is known. Callus cultured cells can be appropriately used to transform plant cells.


Furthermore, the following cells can be used as fungal cells:


yeasts: the Saccharomyces genus such as Saccharomyces serevisiae, and the Pichia genus such as Pichia pastoris; and filamentous fungi: the Aspergillus genus such as Aspergillus niger.


Furthermore, antibody gene expression systems that utilize prokaryotic cells are also known. For example, when using bacterial cells, E. coli cells, Bacillus subtilis cells, and such can suitably be utilized in the present invention. Expression vectors carrying the antibody genes of interest are introduced into these cells by transfection. The transfected cells are cultured in vitro, and the desired antibody can be prepared from the culture of transformed cells.


In addition to the above-described host cells, transgenic animals can also be used to produce a recombinant antibody. That is, the antibody can be obtained from an animal into which the gene encoding the antibody of interest is introduced. For example, the antibody gene can be constructed as a fusion gene by inserting in frame into a gene that encodes a protein produced specifically in milk. Goat β-casein or such can be used, for example, as the protein secreted in milk. DNA fragments containing the fused gene inserted with the antibody gene is injected into a goat embryo, and then this embryo is introduced into a female goat. Desired antibodies can be obtained as a protein fused with the milk protein from milk produced by the transgenic goat born from the embryo-recipient goat (or progeny thereof). In addition, to increase the volume of milk containing the desired antibody produced by the transgenic goat, hormones can be administered to the transgenic goat as necessary (Ebert, K. M. et al., Bio/Technology (1994) 12(7): 699-702).


When an antigen-binding molecule described herein is administered to human, an antigen-binding domain derived from a genetically recombinant antibody that has been artificially modified to reduce the heterologous antigenicity against human and such, can be appropriately used as the antigen-binding domain of the antigen-binding molecule. Such genetically recombinant antibodies include, for example, humanized antibodies. These modified antibodies are appropriately produced by known methods.


An antibody variable region used to produce the antigen-binding domain of an antigen-binding molecule described herein is generally formed by three complementarity-determining regions (CDRs) that are separated by four framework regions (FRs). CDR is a region that substantially determines the binding specificity of an antibody. The amino acid sequences of CDRs are highly diverse. On the other hand, the FR-forming amino acid sequences often have high identity even among antibodies with different binding specificities. Therefore, generally, the binding specificity of a certain antibody can be introduced to another antibody by CDR grafting.


A humanized antibody is also called a reshaped human antibody. Specifically, humanized antibodies prepared by applying the CDR grafting technology, which grafts the CDRs of a non-human animal antibody such as a mouse antibody to a human antibody, and such are known. Common genetic engineering techniques for obtaining humanized antibodies are also known. Specifically, for example, overlap extension PCR is known as a method for grafting a mouse antibody CDR to a human FR. In overlap extension PCR, a nucleotide sequence encoding a mouse antibody CDR to be grafted is added to primers for synthesizing a human antibody FR. Primers are prepared for each of the four FRs. It is generally considered that when grafting a mouse CDR to a human FR, selecting a human FR that has high identity to a mouse FR is advantageous for maintaining the CDR function. That is, it is generally preferable to use a human FR comprising an amino acid sequence which has high identity to the amino acid sequence of the FR adjacent to the mouse CDR to be grafted.


Nucleotide sequences to be ligated are designed so that they will be connected to each other in frame. Human FRs are individually synthesized using the respective primers. As a result, products in which the mouse CDR-encoding DNA is attached to the individual FR-encoding DNAs are obtained. Nucleotide sequences encoding the mouse CDR of each product are designed so that they overlap with each other. Then, complementary strand synthesis reaction is conducted to anneal the overlapping CDR regions of the products synthesized using a human antibody gene as template. Human FRs are ligated via the mouse CDR sequences by this reaction.


The full length V region gene, in which three CDRs and four FRs are ultimately ligated, is amplified using primers that anneal to its 5′- or 3′-end, which are added with suitable restriction enzyme recognition sequences. An expression vector for humanized antibody can be produced by inserting the DNA obtained as described above and a DNA that encodes a human antibody C region into an expression vector so that they will ligate in frame. After the recombinant vector is transfected into a host to establish recombinant cells, the recombinant cells are cultured, and the DNA encoding the humanized antibody is expressed to produce the humanized antibody in the cell culture (see, European Patent Publication No. EP 239400 and International Patent Publication No. WO 1996/002576).


By qualitatively or quantitatively measuring and evaluating the antigen-binding activity of the humanized antibody produced as described above, one can suitably select human antibody FRs that allow CDRs to form a favorable antigen-binding site when ligated through the CDRs. Amino acid residues in FRs may be substituted as necessary, so that the CDRs of a reshaped human antibody form an appropriate antigen-binding site. For example, amino acid sequence mutations can be introduced into FRs by applying the PCR method used for grafting a mouse CDR into a human FR. More specifically, partial nucleotide sequence mutations can be introduced into primers that anneal to the FR. Nucleotide sequence mutations are introduced into the FRs synthesized by using such primers. Mutant FR sequences having the desired characteristics can be selected by measuring and evaluating the activity of the amino acid-substituted mutant antibody to bind to the antigen by the above-mentioned method (Cancer Res. (1993) 53: 851-856).


Alternatively, desired human antibodies can be obtained by immunizing transgenic animals having the entire repertoire of human antibody genes (see International Publication Nos. WO 1993/012227; WO 1992/003918; WO 1994/002602; WO 1994/025585; WO 1996/034096; WO 1996/033735) by DNA immunization.


Furthermore, techniques for preparing human antibodies by panning using human antibody libraries are also known. For example, the V region of a human antibody is expressed as a single-chain antibody (scFv) on phage surface by the phage display method. Phages expressing an scFv that binds to the antigen can be selected. The DNA sequence encoding the human antibody V region that binds to the antigen can be determined by analyzing the genes of selected phages. The DNA sequence of the scFv that binds to the antigen is determined. An expression vector is prepared by fusing the V region sequence in frame with the C region sequence of a desired human antibody, and inserting this into an appropriate expression vector. The expression vector is introduced into cells appropriate for expression such as those described above. The human antibody can be produced by expressing the human antibody-encoding gene in the cells. These methods are already known (see International Publication Nos. WO 1992/001047; WO 1992/020791; WO 1993/006213; WO 1993/011236; WO 1993/019172; WO 1995/001438; WO 1995/015388).


In addition to the techniques described above, techniques of B cell cloning (identification of each antibody-encoding sequence, cloning and its isolation; use in constructing expression vector in order to prepare each antibody (IgG1, IgG2, IgG3, or IgG4 in particular); and such) such as described in Bernasconi et al. (Science (2002) 298: 2199-2202) or in International Publication No. WO 2008/081008 can be appropriately used to isolate antibody genes.


EU Numbering and Kabat Numbering

According to the methods used in the present invention, amino acid positions assigned to antibody CDR and FR are specified according to Kabat's numbering (Sequences of Proteins of Immunological Interest (National Institute of Health, Bethesda, Md., 1987 and 1991)). Herein, when an antigen-binding molecule is an antibody or antigen-binding fragment, variable region amino acids are indicated according to Kabat's numbering system (Kabat numbering), while constant region amino acids are indicated according to EU numbering system based on Kabat's amino acid positions.


Conditions of Ion Concentration
Conditions of Metal Ion Concentration

In one embodiment of the present invention, the ion concentration refers to a metal ion concentration. “Metal ions” refer to ions of group I elements except hydrogen such as alkaline metals and copper group elements, group II elements such as alkaline earth metals and zinc group elements, group III elements except boron, group IV elements except carbon and silicon, group VIII elements such as iron group and platinum group elements, elements belonging to subgroup A of groups V, VI, and VII, and metal elements such as antimony, bismuth, and polonium. Metal atoms have the property of releasing valence electrons to become cations. This is referred to as ionization tendency. Metals with strong ionization tendency are deemed to be chemically active.


In the present invention, preferred metal ions include, for example, calcium ion. Calcium ion is involved in modulation of many biological phenomena, including contraction of muscles such as skeletal, smooth, and cardiac muscles; activation of movement, phagocytosis, and the like of leukocytes; activation of shape change, secretion, and the like of platelets; activation of lymphocytes; activation of mast cells including secretion of histamine; cell responses mediated by catecholamine a receptor or acetylcholine receptor; exocytosis; release of transmitter substances from neuron terminals; and axoplasmic flow in neurons. Known intracellular calcium ion receptors include troponin C, calmodulin, parvalbumin, and myosin light chain, which have several calcium ion-binding sites and are believed to be derived from a common origin in terms of molecular evolution. There are also many known calcium-binding motifs. Such well-known motifs include, for example, cadherin domains, EF-hand of calmodulin, C2 domain of Protein kinase C, Gla domain of blood coagulation protein Factor IX, C-type lectins of asialoglycoprotein receptor and mannose-binding receptor, A domains of LDL receptors, annexin, thrombospondin type 3 domain, and EGF-like domains.


In the present invention, when the metal ion is calcium ion, the conditions of calcium ion concentration include low calcium ion concentration conditions and high calcium ion concentration conditions. “The binding activity varies depending on calcium ion concentration conditions” means that the antigen-binding activity of an antigen-binding molecule varies due to the difference in the conditions between low and high calcium ion concentration conditions. For example, the antigen-binding activity of an antigen-binding molecule may be higher under a high calcium ion concentration condition than under a low calcium ion concentration condition. Alternatively, the antigen-binding activity of an antigen-binding molecule may be higher under a low calcium ion concentration condition than under a high calcium ion concentration condition.


Herein, the high calcium ion concentration is not particularly limited to a specific value; however, the concentration may preferably be selected between 100 μM and 10 mM. In another embodiment, the concentration may be selected between 200 μM and 5 mM. In an alternative embodiment, the concentration may be selected between 400 μM and 3 mM. In still another embodiment, the concentration may be selected between 200 μM and 2 mM. Furthermore, the concentration may be selected between 400 μM and 1 mM. In particular, a concentration selected between 500 μM and 2.5 mM, which is close to the plasma (blood) concentration of calcium ion in vivo, is preferred.


Herein, the low calcium ion concentration is not particularly limited to a specific value; however, the concentration may preferably be selected between 0.1 μM and 30 In another embodiment, the concentration may be selected between 0.2 μM and 20 In still another embodiment, the concentration may be selected between 0.5 μM and 10 In an alternative embodiment, the concentration may be selected between 1 μM and 5 Furthermore, the concentration may be selected between 2 μM and 4 In particular, a concentration selected between 1 μM and 5 which is close to the concentration of ionized calcium in early endosomes in vivo, is preferred.


In the present invention, “the antigen-binding activity is lower at a low calcium ion concentration condition than at a high calcium ion concentration condition” means that the antigen-binding activity of an antigen-binding molecule is weaker at a calcium ion concentration selected between 0.1 μM and 30 μM than at a calcium ion concentration selected between 100 μM and 10 mM. Preferably, it means that the antigen-binding activity of an antigen-binding molecule is weaker at a calcium ion concentration selected between 0.5 μM and 10 μM than at a calcium ion concentration selected between 200 μM and 5 mM. It particularly preferably means that the antigen-binding activity at the calcium ion concentration in the early endosome in vivo is weaker than that at the in vivo plasma calcium ion concentration; and specifically, it means that the antigen-binding activity of an antigen-binding molecule is weaker at a calcium ion concentration selected between 1 μM and 5 μM than at a calcium ion concentration selected between 500 μM and 2.5 mM.


Whether the antigen-binding activity of an antigen-binding molecule is changed depending on metal ion concentration conditions can be determined, for example, by the use of known measurement methods such as those described in the section “Binding Activity” above. For example, in order to confirm that the antigen-binding activity of an antigen-binding molecule becomes higher under a high calcium ion concentration condition than under a low calcium ion concentration condition, the antigen-binding activity of the antigen-binding molecule under low and high calcium ion concentration conditions is compared.


In the present invention, the expression “the antigen-binding activity is lower at a low calcium ion concentration condition than at a high calcium ion concentration condition” can also be expressed as “the antigen-binding activity of an antigen-binding molecule is higher under a high calcium ion concentration condition than under a low calcium ion concentration condition”. In the present invention, “the antigen-binding activity is lower at a low calcium ion concentration condition than at a high calcium ion concentration condition” is sometimes written as “the antigen-binding ability is weaker under a low calcium ion concentration condition than under a high calcium ion concentration condition”. Also, “the antigen-binding activity at a low calcium ion concentration condition is reduced to be lower than that at a high calcium ion concentration condition” may be written as “the antigen-binding ability under a low calcium ion concentration condition is made weaker than that under a high calcium ion concentration condition”.


When determining the antigen-binding activity, the conditions other than calcium ion concentration can be appropriately selected by those skilled in the art, and are not particularly limited. For example, the activity can be determined at 37° C. in HEPES buffer. For example, Biacore (GE Healthcare) or such can be used for the determination. When the antigen is a soluble antigen, the antigen-binding activity of an antigen-binding molecule can be assessed by flowing the antigen as an analyte over a chip onto which the antigen-binding molecule is immobilized. When the antigen is a membrane antigen, the binding activity of an antigen-binding molecule to the membrane antigen can be assessed by flowing the antigen-binding molecule as an analyte over a chip onto which the antigen is immobilized.


As long as the antigen-binding activity of an antigen-binding molecule of the present invention is weaker at a low calcium ion concentration condition than at a high calcium ion concentration condition, the ratio of the antigen-binding activity between under low and high calcium ion concentration conditions is not particularly limited. However, the ratio of the KD (dissociation constant) of the antigen-binding molecule for an antigen at a low calcium ion concentration condition with respect to the KD at a high calcium ion concentration condition, i.e., the value of KD (3 μM Ca)/KD (2 mM Ca), is preferably 2 or more, more preferably 10 or more, and still more preferably 40 or more. The upper limit of the KD (3 μM Ca)/KD (2 mM Ca) value is not particularly limited, and may be any value such as 400, 1000, or 10000 as long as the molecule can be produced by techniques known to those skilled in the art.


When the antigen is a soluble antigen, KD (dissociation constant) can be used to represent the antigen-binding activity. Meanwhile, when the antigen is a membrane antigen, apparent KD (apparent dissociation constant) can be used to represent the activity. KD (dissociation constant) and apparent KD (apparent dissociation constant) can be determined by methods known to those skilled in the art, for example, using Biacore (GE healthcare), Scatchard plot, or flow cytometer.


Alternatively, for example, the dissociation rate constant (kd) can also be preferably used as an index to represent the ratio of the antigen-binding activity of an antigen-binding molecule of the present invention between low and high calcium concentrations. When the dissociation rate constant (kd) is used instead of the dissociation constant (1(D) as an index to represent the binding activity ratio, the ratio of the dissociation rate constant (kd) between low and high calcium concentration conditions, i.e., the value of kd (low calcium concentration condition)/kd (high calcium concentration condition), is preferably 2 or more, more preferably 5 or more, still more preferably 10 or more, and yet more preferably 30 or more. The upper limit of the Kd (low calcium concentration condition)/kd (high calcium concentration condition) value is not particularly limited, and can be any value such as 50, 100, or 200 as long as the molecule can be produced by techniques known to those skilled in the art.


When the antigen is a soluble antigen, kd (dissociation rate constant) can be used to represent the antigen-binding activity. Meanwhile, when the antigen is a membrane antigen, apparent kd (apparent dissociation rate constant) can be used to represent the antigen-binding activity. The kd (dissociation rate constant) and apparent kd (apparent dissociation rate constant) can be determined by methods known to those skilled in the art, for example, using Biacore (GE healthcare) or flow cytometer. In the present invention, when the antigen-binding activity of an antigen-binding molecule is determined at different calcium ion concentrations, it is preferable to use the same conditions except for the calcium concentrations.


For example, an antigen-binding domain (or antigen-binding molecule) whose antigen-binding activity is lower at a low calcium ion concentration condition than at a high calcium ion concentration condition, which is one embodiment of the present invention, can be obtained via screening of antigen-binding domains (or antigen-binding molecules) including the steps of (a) to (c) below:

  • (a) determining the antigen-binding activity of an antigen-binding domain (or antigen-binding molecule) at a low calcium concentration condition;
  • (b) determining the antigen-binding activity of an antigen-binding domain (or antigen-binding molecule) at a high calcium concentration condition; and
  • (c) selecting an antigen-binding domain (or antigen-binding molecule) whose antigen-binding activity is lower at a low calcium concentration condition than at a high calcium concentration condition.


Moreover, an antigen-binding domain (or antigen-binding molecule) whose antigen-binding activity is lower at a low calcium ion concentration condition than at a high calcium ion concentration condition, which is one embodiment of the present invention, can be obtained via screening of antigen-binding domains (or antigen-binding molecules) or a library thereof, including the steps of (a) to (c) below:

  • (a) contacting an antigen with an antigen-binding domain (or antigen-binding molecule), or a library thereof at a high calcium concentration condition;
  • (b) incubating under a low calcium concentration condition an antigen-binding domain (or antigen-binding molecule) that has bound to the antigen in step (a); and
  • (c) isolating an antigen-binding domain (or antigen-binding molecule) dissociated in step (b).


Furthermore, an antigen-binding domain (or antigen-binding molecule) whose antigen-binding activity is lower at a low calcium ion concentration condition than at a high calcium ion concentration condition, which is one embodiment of the present invention, can be obtained via screening of antigen-binding domains (or antigen-binding molecules) or a library thereof, including the steps of (a) to (d) below:

  • (a) contacting an antigen with a library of antigen-binding domains (or antigen-binding molecules) under a low calcium concentration condition;
  • (b) selecting an antigen-binding domain (or antigen-binding molecule) which does not bind to the antigen in step (a);
  • (c) allowing the antigen-binding domain (or antigen-binding molecule) selected in step (b) to bind to the antigen under a high calcium concentration condition; and
  • (d) isolating an antigen-binding domain (or antigen-binding molecule) that has bound to the antigen in step (c).


In addition, an antigen-binding domain (or antigen-binding molecule) whose antigen-binding activity is lower at a low calcium ion concentration condition than at a high calcium ion concentration condition, which is one embodiment of the present invention, can be obtained by a screening method comprising the steps of (a) to (c) below:

  • (a) contacting under a high calcium concentration condition a library of antigen-binding domains (or antigen-binding molecules) with a column onto which an antigen is immobilized;
  • (b) eluting an antigen-binding domain (or antigen-binding molecule) that has bound to the column in step (a) from the column under a low calcium concentration condition; and
  • (c) isolating the antigen-binding domain (or antigen-binding molecule) eluted in step (b).


Furthermore, an antigen-binding domain (or antigen-binding molecule) whose antigen-binding activity is lower at a low calcium ion concentration condition than at a high calcium ion concentration condition, which is one embodiment of the present invention, can be obtained by a screening method comprising the steps of (a) to (d) below:

  • (a) allowing under a low calcium concentration condition a library of antigen-binding domains (or antigen-binding molecules) to pass through a column onto which an antigen is immobilized;
  • (b) collecting an antigen-binding domain (or antigen-binding molecule) that has been eluted without binding to the column in step (a);
  • (c) allowing the antigen-binding domain (or antigen-binding molecule) collected in step (b) to bind to the antigen under a high calcium concentration condition; and
  • (d) isolating an antigen-binding domain (or antigen-binding molecule) that has bound to the antigen in step (c).


Moreover, an antigen-binding domain (or antigen-binding molecule) whose antigen-binding activity is lower at a low calcium ion concentration condition than at a high calcium ion concentration condition, which is one embodiment of the present invention, can be obtained by a screening method comprising the steps of (a) to (d) below:

  • (a) contacting an antigen with a library of antigen-binding domains (or antigen-binding molecules) under a high calcium concentration condition;
  • (b) obtaining an antigen-binding domain (or antigen-binding molecule) that has bound to the antigen in step (a);
  • (c) incubating under a low calcium concentration condition the antigen-binding domain (or antigen-binding molecule) obtained in step (b); and
  • (d) isolating an antigen-binding domain (or antigen-binding molecule) whose antigen-binding activity in step (c) is weaker than the criterion for the selection of step (b).


The above-described steps may be repeated twice or more times. Thus, the present invention provides antigen-binding domains (or antigen-binding molecules) whose antigen-binding activity is lower at a low calcium ion concentration condition than at a high calcium ion concentration condition, which are obtained by screening methods that further comprises the step of repeating twice or more times steps (a) to (c) or (a) to (d) in the above-described screening methods. The number of cycles of steps (a) to (c) or (a) to (d) is not particularly limited, but generally is 10 or less.


In the screening methods of the present invention, the antigen-binding activity of an antigen-binding domain (or antigen-binding molecule) under a low calcium concentration condition is not particularly limited as long as it is antigen-binding activity at an ionized calcium concentration of between 0.1 μM and 30 but preferably is antigen-binding activity at an ionized calcium concentration of between 0.5 μM and 10 More preferably, it is antigen-binding activity at the ionized calcium concentration in the cell in vivo, in particular at the ionized calcium concentration in the early endosome, specifically, between 1 μM and 5 Meanwhile, the antigen-binding activity of an antigen-binding domain (or antigen-binding molecule) under a high calcium concentration condition is not particularly limited, as long as it is antigen-binding activity at an ionized calcium concentration of between 100 μM and 10 mM, but preferably is antigen-binding activity at an ionized calcium concentration of between 200 μM and 5 mM. More preferably, it is antigen-binding activity at the ionized calcium concentration in the cell in vivo, in particular at the ionized calcium concentration in plasma, specifically, between 0.5 mM and 2.5 mM.


The antigen-binding activity of an antigen-binding domain (or antigen-binding molecule) can be measured by methods known to those skilled in the art. Conditions other than the ionized calcium concentration can be determined by those skilled in the art. The antigen-binding activity of an antigen-binding domain (or antigen-binding molecule) can be evaluated as a dissociation constant (1(D), apparent dissociation constant (apparent KD), dissociation rate constant (kd), apparent dissociation constant (apparent kd), and such. These can be determined by methods known to those skilled in the art, for example, using Biacore (GE healthcare), Scatchard plot, or FACS.


In the present invention, the step of selecting an antigen-binding domain (or antigen-binding molecule) whose antigen-binding activity is higher under a high calcium concentration condition than under a low calcium concentration condition is synonymous with the step of selecting an antigen-binding domain (or antigen-binding molecule) whose antigen-binding activity is lower under a low calcium concentration condition than under a high calcium concentration condition.


As long as the antigen-binding activity is higher under a high calcium concentration condition than under a low calcium concentration condition, the difference in the antigen-binding activity between high and low calcium concentration conditions is not particularly limited; however, the antigen-binding activity under a high calcium concentration condition is preferably twice or more, more preferably 10 times or more, and still more preferably 40 times or more than that under a low calcium concentration condition.


Antigen-binding domains (or antigen-binding molecules) of the present invention to be screened by the screening methods described above may be any antigen-binding domains (or antigen-binding molecules). For example, it is possible to screen the above-described antigen-binding domains (or antigen-binding molecules). For example, antigen-binding domains (or antigen-binding molecules) having natural sequences or substituted amino acid sequences may be screened.


Libraries (Library)

In an embodiment, an antigen-binding domain (or antigen-binding molecule) of the present invention can be obtained from a library that is mainly composed of a plurality of antigen-binding molecules whose sequences are different from one another and whose antigen-binding domains have at least one amino acid residue that alters the antigen-binding activity of the antigen-binding molecules depending on ion concentration conditions. The ion concentrations preferably include, for example, metal ion concentration and proton concentration.


Herein, a “library” refers to a plurality of antigen-binding molecules or a plurality of fusion polypeptides containing antigen-binding molecules, or nucleic acids or polynucleotides encoding their sequences. The sequences of a plurality of antigen-binding molecules or a plurality of fusion polypeptides containing antigen-binding molecules in a library are not identical, but are different from one another.


Herein, the phrase “sequences are different from one another” in the expression “a plurality of antigen-binding molecules whose sequences are different from one another” means that the sequences of antigen-binding molecules in a library are different from one another. Specifically, in a library, the number of sequences different from one another reflects the number of independent clones with different sequences, and may also be referred to as “library size”. The library size of a conventional phage display library ranges from 106 to 1012. The library size can be increased up to 1014 by the use of known techniques such as ribosome display. However, the actual number of phage particles used in panning selection of a phage library is in general 10-10000 times greater than the library size. This excess multiplicity is also referred to as “the number of library equivalents”, and means that there are 10 to 10000 individual clones that have the same amino acid sequence. Thus, in the present invention, the phrase “sequences are different from one another” means that the sequences of independent antigen-binding molecules in a library, excluding library equivalents, are different from one another. More specifically, the above means that there are 106 to 1014 antigen-binding molecules whose sequences are different from one another, preferably 107 to 1012 molecules, more preferably 108 to 1011 molecules, and particularly preferably 108 to 1010 molecules whose sequences are different from one another.


In the present invention, the phrase “a plurality of” in the expression “a library mainly composed of a plurality of antigen-binding molecules” generally refers to, in the case of, for example, antigen-binding molecules, fusion polypeptides, polynucleotide molecules, vectors, or viruses of the present invention, a group of two or more types of the substance. For example, when two or more substances are different from one another in a particular characteristic, this means that there are two or more types of the substance. Such examples may include, for example, mutant amino acids observed at specific amino acid positions in an amino acid sequence. For example, when there are two or more antigen-binding molecules of the present invention whose sequences are substantially the same or preferably the same except for flexible residues or except for particular mutant amino acids at hypervariable positions exposed on the surface, there are a plurality of antigen-binding molecules of the present invention. In another Example, when there are two or more polynucleotide molecules whose sequences are substantially the same or preferably the same except for nucleotides encoding flexible residues or nucleotides encoding mutant amino acids of hypervariable positions exposed on the surface, there are a plurality of polynucleotide molecules of the present invention.


In addition, in the present invention, the phrase “mainly composed of” in the expression “a library mainly composed of a plurality of antigen-binding molecules” reflects the number of antigen-binding molecules whose antigen-binding activity varies depending on ion concentration conditions, among independent clones with different sequences in a library. Specifically, it is preferable that there are at least 104 antigen-binding molecules having such binding activity in a library. More preferably, antigen-binding domains of the present invention can be obtained from a library containing at least 105 antigen-binding molecules having such binding activity. Still more preferably, antigen-binding domains of the present invention can be obtained from a library containing at least 106 antigen-binding molecules having such binding activity. Particularly preferably, antigen-binding domains of the present invention can be obtained from a library containing at least 107 antigen-binding molecules having such binding activity. Yet more preferably, antigen-binding domains of the present invention can be obtained from a library containing at least 108 antigen-binding molecules having such binding activity. Alternatively, this may also be preferably expressed as the ratio of the number of antigen-binding molecules whose antigen-binding activity varies depending on ion concentration conditions with respect to the number of independent clones having different sequences in a library. Specifically, antigen-binding domains of the present invention can be obtained from a library in which antigen-binding molecules having such binding activity account for 0.1% to 80%, preferably 0.5% to 60%, more preferably 1% to 40%, still more preferably 2% to 20%, and particularly preferably 4% to 10% of independent clones with different sequences in the library. In the case of fusion polypeptides, polynucleotide molecules, or vectors, similar expressions may be possible using the number of molecules or the ratio to the total number of molecules. In the case of viruses, similar expressions may also be possible using the number of virions or the ratio to total number of virions.


Amino Acids that Alter the Antigen-Binding Activity of Antigen-Binding Domains Depending on Calcium Ion Concentration Conditions


Antigen-binding domains (or antigen-binding molecules) of the present invention to be screened by the above-described screening methods may be prepared in any manner. For example, when the metal ion is calcium ion, it is possible to use preexisting antibodies, preexisting libraries (phage library, etc.), antibodies or libraries prepared from hybridomas obtained by immunizing animals or from B cells of immunized animals, antibodies or libraries obtained by introducing amino acids capable of chelating calcium (for example, aspartic acid and glutamic acid) or unnatural amino acid mutations into the above-described antibodies or libraries (calcium-chelatable amino acids (such as aspartic acid and glutamic acid), libraries with increased content of unnatural amino acids, libraries prepared by introducing calcium-chelatable amino acids (such as aspartic acid and glutamic acid) or unnatural amino acid mutations at particular positions, or the like.


Examples of the amino acids that alter the antigen-binding activity of antigen-binding molecules depending on ion concentration conditions as described above may be any types of amino acids as long as the amino acids form a calcium-binding motif. Calcium-binding motifs are well known to those skilled in the art and have been described in details (for example, Springer et al. (Cell (2000) 102, 275-277); Kawasaki and Kretsinger (Protein Prof. (1995) 2, 305-490); Moncrief et al. (J. Mol. Evol. (1990) 30, 522-562); Chauvaux et al. (Biochem. J. (1990) 265, 261-265); Bairoch and Cox (FEBS Lett. (1990) 269, 454-456); Davis (New Biol. (1990) 2, 410-419); Schaefer et al. (Genomics (1995) 25, 638-643); Economou et al. (EMBO J. (1990) 9, 349-354); Wurzburg et al. (Structure. (2006) 14, 6, 1049-1058)). Specifically, any known calcium-binding motifs, including type C lectins such as ASGPR, CD23, MBR, and DC-SIGN, can be included in antigen-binding molecules of the present invention. Preferred examples of such preferred calcium-binding motifs also include, in addition to those described above, for example, the calcium-binding motif in the antigen-binding domain of SEQ ID NO: 2.


Furthermore, as amino acids that alter the antigen-binding activity of antigen-binding molecules depending on calcium ion concentration conditions, for example, amino acids having metal-chelating activity may also be preferably used. Examples of such metal-chelating amino acids include, for example, serine (Ser(S)), threonine (Thr(T)), asparagine (Asn(N)), glutamine (Gln(Q)), aspartic acid (Asp(D)), and glutamic acid (Glu(E)).


Positions in the antigen-binding domains at which the above-described amino acids are contained are not particularly limited to particular positions, and may be any positions within the heavy chain variable region or light chain variable region that forms an antigen-binding domain, as long as they alter the antigen-binding activity of antigen-binding molecules depending on calcium ion concentration conditions. More specifically, antigen-binding domains of the present invention can be obtained from a library mainly composed of antigen-binding molecules whose sequences are different from one another and whose heavy chain antigen-binding domains contain amino acids that alter the antigen-binding activity of the antigen-binding molecules depending on calcium ion concentration conditions. In another embodiment, antigen-binding domains of the present invention can be obtained from a library mainly composed of antigen-binding molecules whose sequences are different from one another and whose heavy chain CDR3 domains contain the above-mentioned amino acids. In still another embodiment, antigen-binding domains of the present invention can be obtained from a library mainly composed of antigen-binding molecules whose sequences are different from one another and whose heavy chain CDR3 domains contain the above-mentioned amino acids at positions 95, 96, 100a, and/or 101 as indicated according to the Kabat numbering system.


Meanwhile, in an embodiment of the present invention, antigen-binding domains of the present invention can be obtained from a library mainly composed of antigen-binding molecules whose sequences are different from one another and whose light chain antigen-binding domains contain amino acids that alter the antigen-binding activity of antigen-binding molecules depending on calcium ion concentration conditions. In another embodiment, antigen-binding domains of the present invention can be obtained from a library mainly composed of antigen-binding molecules whose sequences are different from one another and whose light chain CDR1 domains contain the above-mentioned amino acids. In still another embodiment, antigen-binding domains of the present invention can be obtained from a library mainly composed of antigen-binding molecules whose sequences are different from one another and whose light chain CDR1 domains contain the above-mentioned amino acids at positions 30, 31, and/or 32 as indicated according to the Kabat numbering system.


In another embodiment, antigen-binding domains of the present invention can be obtained from a library mainly composed of antigen-binding molecules whose sequences are different from one another and whose light chain CDR2 domains contain the above-mentioned amino acid residues. In yet another embodiment, the present invention provides libraries mainly composed of antigen-binding molecules whose sequences are different from one another and whose light chain CDR2 domains contain the above-mentioned amino acid residues at position 50 as indicated according to the Kabat numbering system.


In still another embodiment of the present invention, antigen-binding domains of the present invention can be obtained from a library mainly composed of antigen-binding molecules whose sequences are different from one another and whose light chain CDR3 domains contain the above-mentioned amino acid residues. In an alternative embodiment, antigen-binding domains of the present invention can be obtained from a library mainly composed of antigen-binding molecules whose sequences are different from one another and whose light chain CDR3 domains contain the above-mentioned amino acid residues at position 92 as indicated according to the Kabat numbering system.


Furthermore, in a different embodiment of the present invention, antigen-binding domains of the present invention can be obtained from a library mainly composed of antigen-binding molecules whose sequences are different from one another and in which two or three CDRs selected from the above-described light chain CDR1, CDR2, and CDR3 contain the aforementioned amino acid residues. Moreover, antigen-binding domains of the present invention can be obtained from a library mainly composed of antigen-binding molecules whose sequences are different from one another and whose light chains contain the aforementioned amino acid residues at any one or more of positions 30, 31, 32, 50, and/or 92 as indicated according to the Kabat numbering system.


In a particularly preferred embodiment, the framework sequences of the light chain and/or heavy chain variable region of an antigen-binding molecule preferably contain human germ line framework sequences. Thus, in an embodiment of the present invention, when the framework sequences are completely human sequences, it is expected that when such an antigen-binding molecule of the present invention is administered to humans (for example, to treat diseases), it induces little or no immunogenic response. In the above sense, the phrase “containing a germ line sequence” in the present invention means that a part of the framework sequences of the present invention is identical to a part of any human germ line framework sequences. For example, when the heavy chain FR2 sequence of an antigen-binding molecule of the present invention is a combination of heavy chain FR2 sequences of different human germ line framework sequences, such a molecule is also an antigen-binding molecule of the present invention “containing a germ line sequence”.


Preferred examples of the frameworks include, for example, fully human framework region sequences currently known, which are included in the website of V-Base (http://vbase.mrc-cpe.cam.ac.uk/) or others. Those framework region sequences can be appropriately used as a germ line sequence contained in an antigen-binding molecule of the present invention. The germ line sequences may be categorized according to their similarity (Tomlinson et al. (J. Mol. Biol. (1992) 227, 776-798); Williams and Winter (Eur. J. Immunol. (1993) 23, 1456-1461); Cox et al. (Nat. Genetics (1994) 7, 162-168)). Appropriate germ line sequences can be selected from Vκ, which is grouped into seven subgroups; Vλ, which is grouped into ten subgroups; and VH, which is grouped into seven subgroups.


Fully human VH sequences preferably include, but are not limited to, for example, VH sequences of:


subgroup VH1 (for example, VH1-2, VH1-3, VH1-8, VH1-18, VH1-24, VH1-45, VH1-46, VH1-58, and VH1-69);


subgroup VH2 (for example, VH2-5, VH2-26, and VH2-70);


subgroup VH3 (VH3-7, VH3-9, VH3-11, VH3-13, VH3-15, VH3-16, VH3-20, VH3-21, VH3-23, VH3-30, VH3-33, VH3-35, VH3-38, VH3-43, VH3-48, VH3-49, VH3-53, VH3-64, VH3-66, VH3-72, VH3-73, and VH3-74);


subgroup VH4 (VH4-4, VH4-28, VH4-31, VH4-34, VH4-39, VH4-59, and VH4-61);


subgroup VH5 (VH5-51);


subgroup VH6 (VH6-1); and


subgroup VH7 (VH7-4 and VH7-81).


These are also described in known documents (Matsuda et al. (J. Exp. Med. (1998) 188, 1973-1975)) and such, and thus persons skilled in the art can appropriately design antigen-binding molecules of the present invention based on the information of these sequences. It is also preferable to use other fully human frameworks or framework sub-regions.


Fully human VK sequences preferably include, but are not limited to, for example:


A20, A30, L1, L4, L5, L8, L9, L11, L12, L14, L15, L18, L19, L22, L23, L24, O2, O4, O8, O12, O14, and 018 grouped into subgroup Vk1;


A1, A2, A3, A5, A7, A17, A18, A19, A23, 01, and 011, grouped into subgroup Vk2;


A11, A27, L2, L6, L10, L16, L20, and L25, grouped into subgroup Vk3;


B3, grouped into subgroup Vk4;


B2 (herein also referred to as Vk5-2), grouped into subgroup Vk5; and


A10, A14, and A26, grouped into subgroup Vk6 (Kawasaki et al. (Eur. J. Immunol. (2001) 31, 1017-1028); Schable and Zachau (Biol. Chem. Hoppe Seyler (1993) 374, 1001-1022); Brensing-Kuppers et al. (Gene (1997) 191, 173-181)).


Fully human Vλ, sequences preferably include, but are not limited to, for example:


V1-2, V1-3, V1-4, V1-5, V1-7, V1-9, V1-11, V1-13, V1-16, V1-17, V1-18, V1-19, V1-20, and

V1-22, grouped into subgroup VL1;


V2-1, V2-6, V2-7, V2-8, V2-11, V2-13, V2-14, V2-15, V2-17, and V2-19, grouped into subgroup VL1;


V3-2, V3-3, and V3-4, grouped into subgroup VL3;


V4-1, V4-2, V4-3, V4-4, and V4-6, grouped into subgroup VL4; and


V5-1, V5-2, V5-4, and V5-6, grouped into subgroup VL5 (Kawasaki et al. (Genome Res. (1997) 7, 250-261)).


Normally, these framework sequences are different from one another at one or more amino acid residues. These framework sequences can be used in combination with “at least one amino acid residue that alters the antigen-binding activity of an antigen-binding molecule depending on ion concentration conditions” of the present invention. Other examples of the fully human frameworks used in combination with “at least one amino acid residue that alters the antigen-binding activity of an antigen-binding molecule depending on ion concentration conditions” of the present invention include, but are not limited to, for example, KOL, NEWM, REI, EU, TUR, TEI, LAY, and POM (for example, Kabat et al. (1991) supra; Wu et al. (J. Exp. Med. (1970) 132, 211-250)).


Without being bound by a particular theory, one reason for the expectation that the use of germ line sequences precludes adverse immune responses in most individuals is believed to be as follows. As a result of the process of affinity maturation during normal immune responses, somatic mutation occurs frequently in the variable regions of immunoglobulin. Such mutations mostly occur around CDRs whose sequences are hypervariable, but also affect residues of framework regions. Such framework mutations do not exist on the germ line genes, and also they are less likely to be immunogenic in patients. On the other hand, the normal human population is exposed to most of the framework sequences expressed from the germ line genes. As a result of immunotolerance, these germ line frameworks are expected to have low or no immunogenicity in patients. To maximize the possibility of immunotolerance, variable region-encoding genes may be selected from a group of commonly occurring functional germ line genes.


Known methods such as site-directed mutagenesis (Kunkel et al. (Proc. Natl. Acad. Sci. USA (1985) 82, 488-492)) and overlap extension PCR can be appropriately employed to produce the antigen-binding molecules of the present invention in which the above-described framework sequences contain amino acids that alter the antigen-binding activity of the antigen-binding molecules depending on calcium ion concentration conditions.


For example, a library which contains a plurality of antigen-binding molecules of the present invention whose sequences are different from one another can be constructed by combining heavy chain variable regions prepared as a randomized variable region sequence library with a light chain variable region selected as a framework sequence originally containing at least one amino acid residue that alters the antigen-binding activity of the antigen-binding molecule depending on calcium ion concentration conditions. As a non-limiting example, when the ion concentration is calcium ion concentration, such preferred libraries include, for example, those constructed by combining the light chain variable region sequence of SEQ ID NO: 2 (Vk5-2) and the heavy chain variable region produced as a randomized variable region sequence library.


Alternatively, a light chain variable region sequence selected as a framework region originally containing at least one amino acid residue that alters the antigen-binding activity of an antigen-binding molecule depending on ion concentration conditions as mentioned above can be designed to contain various amino acid residues other than the above amino acid residues. In the present invention, such residues are referred to as flexible residues. The number and position of flexible residues are not particularly limited as long as the antigen-binding activity of the antigen-binding molecule of the present invention varies depending on ion concentration conditions. Specifically, the CDR sequences and/or FR sequences of the heavy chain and/or light chain may contain one or more flexible residues. For example, when the ion concentration is calcium ion concentration, non-limiting examples of flexible residues to be introduced into the light chain variable region sequence of SEQ ID NO: 2 (Vk5-2) include the amino acid residues listed in Table 1 or 2.











TABLE 1





CDR
Position
70% of the total





















CDR1
28
S; 100%







29
I; 100%



30
E; 72%
H; 14%
S; 14%



31
D; 100%



32
D; 100%



33
L; 100%



34
A; 70%
N; 30%


CDR2
50
E; 100%



51
A; 100%



52
S; 100%



53
H; 5%
N; 25%
S; 45%
T; 25%



54
L; 100%



55
Q; 100%



56
S; 100%


CDR3
90
Q; 100%



91
H; 25%
S; 15%
R; 15%
Y; 45%



92
D; 80%
N; 10%
S; 10%



93
D; 5%
G; 10%
N; 25%
S; 50%
R; 10%



94
S; 50%
Y; 50%



95
P; 100%



96
L; 50%
Y; 50%





(Position indicates Kabat numbering)






When position 92 as indicated by Kabat numbering is Asn (N), position 94 may be Leu (L) rather than Ser (S).











TABLE 2






Kabat



CDR
numbering
30% amino acids of the total





















CDR1
28
S: 100%







29
I: 100%



30
E: 83%
S: 17%



31
D: 100%



32
D: 100%



33
L: 100%



34
A: 70%
N: 30%


CDR2
50
H: 100%



51
A: 100%



52
S: 100%



53
H: 5%
N: 25%
S: 45%
T: 25%



54
L: 100%



55
Q: 100%



56
S: 100%


CDR3
90
Q: 100%



91
H: 25%
S: 15%
R: 15%
Y: 45%



92
D: 80%
N: 10%
S: 10%



93
D: 5%
G: 10%
N: 25%
S: 50%
R: 10%



94
S: 50%
Y: 50%



95
P: 100%



96
L: 50%
Y: 50%





(Position indicates Kabat numbering)






When position 92 as indicated by Kabat numbering is Asn (N), position 94 may be Leu (L) rather than Ser (S).


Herein, flexible residues refer to amino acid residue variations present at hypervariable positions at which several different amino acids are present on the light chain and heavy chain variable regions when the amino acid sequences of known and/or native antibodies or antigen-binding domains are compared. Hypervariable positions are generally located in the CDR regions. In an embodiment, the data provided by Kabat, Sequences of Proteins of Immunological Interest (National Institute of Health Bethesda Md.) (1987 and 1991) is useful to determine hypervariable positions in known and/or native antibodies. Furthermore, databases on the Internet (http://vbase.mrc-cpe.cam.ac.uk/, http://www.bioinf.org.uk/abs/index.html) provide the collected sequences of many human light chains and heavy chains and their locations. The information on the sequences and locations is useful to determine hypervariable positions in the present invention. According to the present invention, when a certain amino acid position has preferably about 2 to about 20 possible amino acid residue variations, preferably about 3 to about 19, preferably about 4 to about 18, preferably 5 to 17, preferably 6 to 16, preferably 7 to 15, preferably 8 to 14, preferably 9 to 13, and preferably 10 to 12 possible amino acid residue variations, the position is hypervariable. In some embodiments, a certain amino acid position may have preferably at least about 2, preferably at least about 4, preferably at least about 6, preferably at least about 8, preferably about 10, and preferably about 12 amino acid residue variations.


Alternatively, a library containing a plurality of antigen-binding molecules of the present invention whose sequences are different from one another can be constructed by combining heavy chain variable regions produced as a randomized variable region sequence library with light chain variable regions into which at least one amino acid residue that alters the antigen-binding activity of antigen-binding molecules depending on ion concentration conditions as mentioned above is introduced. When the ion concentration is calcium ion concentration, non-limiting examples of such libraries preferably include, for example, libraries in which heavy chain variable regions produced as a randomized variable region sequence library are combined with light chain variable region sequences in which a particular residue(s) in a germ line sequence such as SEQ ID NO: 3 (Vk1), SEQ ID NO: 4 (Vk2), SEQ ID NO: 5 (Vk3), or SEQ ID NO: 6 (Vk4) has been substituted with at least one amino acid residue that alters the antigen-binding activity of an antigen-binding molecule depending on calcium ion concentration conditions. Non-limiting examples of such amino acid residues include amino acid residues in light chain CDR1. Furthermore, non-limiting examples of such amino acid residues include amino acid residues in light chain CDR2. In addition, non-limiting examples of such amino acid residues also include amino acid residues in light chain CDR3.


Non-limiting examples of such amino acid residues contained in light chain CDR1 include those at positions 30, 31, and/or 32 in the CDR1 of light chain variable region as indicated by EU numbering. Furthermore, non-limiting examples of such amino acid residues contained in light chain CDR2 include an amino acid residue at position 50 in the CDR2 of light chain variable region as indicated by Kabat numbering. Moreover, non-limiting examples of such amino acid residues contained in light chain CDR3 include an amino acid residue at position 92 in the CDR3 of light chain variable region as indicated by Kabat numbering. These amino acid residues can be contained alone or in combination as long as they form a calcium-binding motif and/or as long as the antigen-binding activity of an antigen-binding molecule varies depending on calcium ion concentration conditions. Meanwhile, as troponin C, calmodulin, parvalbumin, and myosin light chain, which have several calcium ion-binding sites and are believed to be derived from a common origin in terms of molecular evolution, are known, the light chain CDR1, CDR2, and/or CDR3 can be designed to have their binding motifs. For example, it is possible to use cadherin domains, EF hand of calmodulin, C2 domain of Protein kinase C, G1a domain of blood coagulation protein FactorIX, C type lectins of asialoglycoprotein receptor and mannose-binding receptor, A domains of LDL receptors, annexin, thrombospondin type 3 domain, and EGF-like domains in an appropriate manner for the above purposes.


When heavy chain variable regions produced as a randomized variable region sequence library and light chain variable regions into which at least one amino acid residue that alters the antigen-binding activity of an antigen-binding molecule depending on ion concentration conditions has been introduced are combined as described above, the sequences of the light chain variable regions can be designed to contain flexible residues in the same manner as described above. The number and position of such flexible residues are not particularly limited to particular embodiments as long as the antigen-binding activity of antigen-binding molecules of the present invention varies depending on ion concentration conditions. Specifically, the CDR sequences and/or FR sequences of heavy chain and/or light chain can contain one or more flexible residues. When the ion concentration is calcium ion concentration, non-limiting examples of flexible residues to be introduced into the sequence of light chain variable region include the amino acid residues listed in Tables 1 and 2.


The preferred heavy chain variable regions to be combined include, for example, randomized variable region libraries. Known methods are combined as appropriate to produce a randomized variable region library. In a non-limiting embodiment of the present invention, an immune library constructed based on antibody genes derived from lymphocytes of animals immunized with a specific antigen, patients with infections, persons with an elevated antibody titer in blood as a result of vaccination, cancer patients, or auto immune disease patients, may be preferably used as a randomized variable region library.


In another non-limiting embodiment of the present invention, a synthetic library produced by replacing the CDR sequences of V genes in genomic DNA or functional reshaped V genes with a set of synthetic oligonucleotides containing sequences encoding codon sets of an appropriate length can also be preferably used as a randomized variable region library. In this case, since sequence diversity is observed in the heavy chain CDR3 sequence, it is also possible to replace the CDR3 sequence only. A criterion of giving rise to diversity in amino acids in the variable region of an antigen-binding molecule is that diversity is given to amino acid residues at surface-exposed positions in the antigen-binding molecule. The surface-exposed position refers to a position that is considered to be able to be exposed on the surface and/or contacted with an antigen, based on structure, ensemble of structures, and/or modeled structure of an antigen-binding molecule. In general, such positions are CDRs. Preferably, surface-exposed positions are determined using coordinates from a three-dimensional model of an antigen-binding molecule using a computer program such as the InsightII program (Accelrys). Surface-exposed positions can be determined using algorithms known in the art (for example, Lee and Richards (J. Mol. Biol. (1971) 55, 379-400); Connolly (J. Appl. Cryst. (1983) 16, 548-558)). Determination of surface-exposed positions can be performed using software suitable for protein modeling and three-dimensional structural information obtained from an antibody. Software that can be used for these purposes preferably includes SYBYL Biopolymer Module software (Tripos Associates). Generally or preferably, when an algorithm requires a user input size parameter, the “size” of a probe which is used in the calculation is set at about 1.4 Angstrom or smaller in radius. Furthermore, methods for determining surface-exposed regions and areas using software for personal computers are described by Pacios (Comput. Chem. (1994) 18 (4), 377-386; J. Mol. Model. (1995) 1, 46-53).


In another non-limiting embodiment of the present invention, a naive library, which is constructed from antibody genes derived from lymphocytes of healthy persons and whose repertoire consists of naive sequences, which are antibody sequences with no bias, can also be particularly preferably used as a randomized variable region library (Gejima et al. (Human Antibodies (2002) 11, 121-129); Cardoso et al. (Scand. J. Immunol. (2000) 51, 337-344)). Herein, an amino acid sequence comprising a naive sequence refers to an amino acid sequence obtained from such a naive library.


In one embodiment of the present invention, an antigen-binding domain of the present invention can be obtained from a library containing a plurality of antigen-binding molecules of the present invention whose sequences are different from one another, prepared by combining light chain variable regions constructed as a randomized variable region sequence library with a heavy chain variable region selected as a framework sequence that originally contains “at least one amino acid residue that alters the antigen-binding activity of an antigen-binding molecule depending on ion concentration conditions”. When the ion concentration is calcium ion concentration, non-limiting examples of such libraries preferably include those constructed by combining light chain variable regions constructed as a randomized variable region sequence library with the sequence of heavy chain variable region of SEQ ID NO: 7 (6RL#9-IgG1) or SEQ ID NO: 8 (6KC4-1#85-IgG1). Alternatively, such a library can be constructed by selecting appropriate light chain variable regions from those having germ line sequences, instead of light chain variable regions constructed as a randomized variable region sequence library. Such preferred libraries include, for example, those in which the sequence of heavy chain variable region of SEQ ID NO: 7 (6RL#9-IgG1) or SEQ ID NO: 8 (6KC4-1#85-IgG1) is combined with light chain variable regions having germ line sequences.


Alternatively, the sequence of a heavy chain variable region selected as a framework sequence that originally contains “at least one amino acid residue that alters the antigen-binding activity of an antigen-binding molecule depending on ion concentration conditions” as mentioned above can be designed to contain flexible residues. The number and position of the flexible residues are not particularly limited as long as the antigen-binding activity of an antigen-binding molecule of the present invention varies depending on ion concentration conditions. Specifically, the CDR and/or FR sequences of heavy chain and/or light chain can contain one or more flexible residues. When the ion concentration is calcium ion concentration, non-limiting examples of flexible residues to be introduced into the sequence of heavy chain variable region of SEQ ID NO: 7 (6RL#9-IgG1) include all amino acid residues of heavy chain CDR1 and CDR2 and the amino acid residues of the heavy chain CDR3 except those at positions 95, 96, and/or 100a. Alternatively, non-limiting examples of flexible residues to be introduced into the sequence of heavy chain variable region of SEQ ID NO: 8 (6KC4-1#85-IgG1) include all amino acid residues of heavy chain CDR1 and CDR2 and the amino acid residues of the heavy chain CDR3 except those at amino acid positions 95 and/or 101.


Alternatively, a library containing a plurality of antigen-binding molecules whose sequences are different from one another can be constructed by combining light chain variable regions constructed as a randomized variable region sequence library or light chain variable regions having germ line sequences with heavy chain variable regions into which “at least one amino acid residue responsible for the ion concentration condition-dependent change in the antigen-binding activity of an antigen-binding molecule” has been introduced as mentioned above. When the ion concentration is calcium ion concentration, non-limiting examples of such libraries preferably include those in which light chain variable regions constructed as a randomized variable region sequence library or light chain variable regions having germ line sequences are combined with the sequence of a heavy chain variable region in which a particular residue(s) has been substituted with at least one amino acid residue that alters the antigen-binding activity of an antigen-binding molecule depending on calcium ion concentration conditions. Non-limiting examples of such amino acid residues include amino acid residues of the heavy chain CDR1. Further non-limiting examples of such amino acid residues include amino acid residues of the heavy chain CDR2. In addition, non-limiting examples of such amino acid residues also include amino acid residues of the heavy chain CDR3. Non-limiting examples of such amino acid residues of heavy chain CDR3 include the amino acids of positions 95, 96, 100a, and/or 101 in the CDR3 of heavy chain variable region as indicated by the Kabat numbering. Furthermore, these amino acid residues can be contained alone or in combination as long as they form a calcium-binding motif and/or the antigen-binding activity of an antigen-binding molecule varies depending on calcium ion concentration conditions.


When light chain variable regions constructed as a randomized variable region sequence library or light chain variable regions having germ line sequence are combined with a heavy chain variable region into which at least one amino acid residue that alter the antigen-binding activity of an antigen-binding molecule depending on ion concentration conditions as mentioned above has been introduced, the sequence of the heavy chain variable region can also be designed to contain flexible residues in the same manner as described above. The number and position of flexible residues are not particularly limited as long as the antigen-binding activity of an antigen-binding molecule of the present invention varies depending on ion concentration conditions. Specifically, the heavy chain CDR and/or FR sequences may contain one or more flexible residues. Furthermore, randomized variable region libraries can be preferably used as amino acid sequences of CDR1, CDR2, and/or CDR3 of the heavy chain variable region other than the amino acid residues that alter the antigen-binding activity of an antigen-binding molecule depending on ion concentration condition. When germ line sequences are used as light chain variable regions, non-limiting examples of such sequences include those of SEQ ID NO: 3 (Vk1), SEQ ID NO: 4 (Vk2), SEQ ID NO: 5 (Vk3), and SEQ ID NO: 6 (Vk4).


Any of the above-described amino acids that alter the antigen-binding activity of an antigen-binding molecule depending on calcium ion concentration conditions can be preferably used, as long as they form a calcium-binding motif. Specifically, such amino acids include electron-donating amino acids. Preferred examples of such electron-donating amino acids include serine, threonine, asparagine, glutamic acid, aspartic acid, and glutamic acid.


Condition of Proton Concentrations

In an embodiment of the present invention, the condition of ion concentrations refers to the condition of proton concentrations or pH condition. In the present invention, the concentration condition of proton, i.e., the nucleus of hydrogen atom, is treated as synonymous with condition of hydrogen index (pH). When the activity of proton in an aqueous solution is represented as aH+, pH is defined as −log 10aH+. When the ionic strength of the aqueous solution is low (for example, lower than 10−3), aH+ is nearly equal to the proton strength. For example, the ionic product of water at 25° C. and 1 atmosphere is Kw=aH+aOH=10−14, and therefore in pure water, aH+=aOH=10−7. In this case, pH=7 is neutral; an aqueous solution whose pH is lower than 7 is acidic or whose pH is greater than 7 is alkaline.


In the present invention, when pH condition is used as the ion concentration condition, pH conditions include condition of high proton concentrations or low pHs, i.e., an acidic pH range condition, and condition of low proton concentrations or high pHs, i.e., a neutral pH range condition. “The binding activity varies depending on pH condition” means that the antigen-binding activity of an antigen-binding molecule varies due to the difference in conditions of a high proton concentration or low pH (an acidic pH range) and a low proton concentration or high pH (a neutral pH range). This includes, for example, the case where the antigen-binding activity of an antigen-binding molecule is higher at a neutral pH range condition than at an acidic pH range condition and the case where the antigen-binding activity of an antigen-binding molecule is higher at an acidic pH range condition than at a neutral pH range condition.


Herein, neutral pH range is not limited to a specific value and is preferably selected from between pH 6.7 and pH 10.0. In another embodiment, the pH can be selected from between pH 6.7 and pH 9.5. In still another embodiment, the pH can be selected from between pH 7.0 and pH 9.0. In yet another embodiment, the pH can be selected from between pH 7.0 and pH 8.0. In particular, the preferred pH includes the extracellular pH in vivo, especially pH 7.4, which is close to the pH of plasma (blood).


Herein, an acidic pH range is not limited to a specific value and is preferably selected from between pH 4.0 and pH 6.5. In another embodiment, the pH can be selected from between pH 4.5 and pH 6.5. In still another embodiment, the pH can be selected from between pH 5.0 and pH 6.5. In yet another embodiment, the pH can be selected from between pH 5.5 and pH 6.5. In particular, the preferred pH includes the extracellular pH in vivo, especially pH 5.8, which is close to the pH in the early endosome in vivo.


In the present invention, “the antigen-binding activity of an antigen-binding molecule at a high proton concentration or low pH (an acidic pH range) condition is lower than that at a low proton concentration or high pH (a neutral pH range) condition” means that the antigen-binding activity of an antigen-binding molecule at a pH selected from between pH 4.0 and pH 6.5 is weaker than that at a pH selected from between pH 6.7 and pH 10.0; preferably means that the antigen-binding activity of an antigen-binding molecule at a pH selected from between pH 4.5 and pH 6.5 is weaker than that at a pH selected from between pH 6.7 and pH 9.5; more preferably, means that the antigen-binding activity of an antigen-binding molecule at a pH selected from between pH 5.0 and pH 6.5 is weaker than that at a pH selected from between pH 7.0 and pH 9.0; still more preferably means that the antigen-binding activity of an antigen-binding molecule at a pH selected from between pH 5.5 and pH 6.5 is weaker than that at a pH selected from between pH 7.0 and pH 8.0; particularly preferably means that the antigen-binding activity at the pH in the early endosome in vivo is weaker than the antigen-binding activity at the pH of plasma in vivo; and specifically means that the antigen-binding activity of an antigen-binding molecule at pH 5.8 is weaker than the antigen-binding activity at pH 7.4.


Whether the antigen-binding activity of an antigen-binding molecule has changed by the pH condition can be determined, for example, by the use of known measurement methods such as those described in the section “Binding Activity” above. Specifically, the binding activity is measured under different pH conditions using the measurement methods described above. For example, the antigen-binding activity of an antigen-binding molecule is compared under the conditions of acidic pH range and neutral pH range to confirm that the antigen-binding activity of the antigen-binding molecule changes to be higher under the condition of neutral pH range than that under the condition of acidic pH range.


Furthermore, in the present invention, the expression “the antigen-binding activity at a condition of high proton concentration or low pH, i.e., in an acidic pH range condition, is lower than that at a condition of low proton concentration or high pH, i.e., in a neutral pH range condition” can also be expressed as “the antigen-binding activity of an antigen-binding molecule at a condition of low proton concentration or high pH, i.e., in a neutral pH range condition, is higher than that at a condition of high proton concentration or low pH, i.e., in an acidic pH range condition”. In the present invention, “the antigen-binding activity at a condition of high proton concentration or low pH, i.e., in an acidic pH range condition, is lower than that at a condition of low proton concentration or high pH, i.e., in a neutral pH range condition” may be described as “the antigen-binding activity at a condition of high proton concentration or low pH, i.e., in an acidic pH range condition, is weaker than the antigen-binding ability at a condition of low proton concentration or high pH, i.e., in a neutral pH range condition”. Alternatively, “the antigen-binding activity at a condition of high proton concentration or low pH, i.e., in an acidic pH range condition, is reduced to be lower than that at a condition of low proton concentration or high pH, i.e., in a neutral pH range condition” may be described as “the antigen-binding activity at a condition of high proton concentration or low pH, i.e., in an acidic pH range condition, is reduced to be weaker than the antigen-binding ability at a condition of low proton concentration or high pH, i.e., in a neutral pH range condition”.


The conditions other than proton concentration or pH for measuring the antigen-binding activity may be suitably selected by those skilled in the art and are not particularly limited. Measurements can be carried out, for example, at 37° C. using HEPES buffer. Measurements can be carried out, for example, using Biacore (GE Healthcare). When the antigen is a soluble antigen, the antigen-binding activity of an antigen-binding molecule can be determined by assessing the binding activity to the soluble antigen by pouring the antigen as an analyte into a chip immobilized with the antigen-binding molecule. When the antigen is a membrane antigen, the binding activity to the membrane antigen can be assessed by pouring the antigen-binding molecule as an analyte into a chip immobilized with the antigen.


As long as the antigen-binding activity of an antigen-binding molecule of the present invention at a condition of high proton concentration or low pH, i.e., in an acidic pH range condition is weaker than that at a condition of low proton concentration or high pH, i.e., in a neutral pH range condition, the ratio of the antigen-binding activity between that under a condition of high proton concentration or low pH, i.e., under an acidic pH range condition, and under a condition of low proton concentration or high pH, i.e., under a neutral pH range condition, is not particularly limited, and the value of KD (pH 5.8)/KD (pH 7.4), which is the ratio of the dissociation constant (KD) for an antigen at a condition of high proton concentration or low pH, i.e., in an acidic pH range condition to the KD at a condition of low proton concentration or high pH, i.e., in a neutral pH range condition, is preferably 2 or more; more preferably the value of KD (pH 5.8)/KD (pH 7.4) is 10 or more; and still more preferably the value of KD (pH 5.8)/KD (pH 7.4) is 40 or more. The upper limit of KD (pH 5.8)/KD (pH 7.4) value is not particularly limited, and may be any value such as 400, 1000, or 10000, as long as the molecule can be produced by the techniques of those skilled in the art.


When the antigen is a soluble antigen, the dissociation constant (1(D) can be used as the value for antigen-binding activity. Meanwhile, when the antigen is a membrane antigen, the apparent dissociation constant (1(D) can be used. The dissociation constant (1(D) and apparent dissociation constant (KD) can be measured by methods known to those skilled in the art, and Biacore (GE healthcare), Scatchard plot, flow cytometer, and such can be used.


Alternatively, for example, the dissociation rate constant (kd) can be suitably used as an index for indicating the ratio of the antigen-binding activity of an antigen-binding molecule of the present invention between that at a condition of high proton concentration or low pH, i.e., an acidic pH range and a condition of low proton concentration or high pH, i.e., a neutral pH range. When kd (dissociation rate constant) is used as an index for indicating the binding activity ratio instead of KD (dissociation constant), the value of kd (in an acidic pH range)/kd (in a neutral pH range), which is the ratio of kd (dissociation rate constant) for the antigen at a condition of high proton concentration or low pH, i.e., in an acidic pH range to kd (dissociation rate constant) at a condition of low proton concentration or high pH, i.e., in a neutral pH range, is preferably 2 or more, more preferably 5 or more, still more preferably 10 or more, and yet more preferably 30 or more. The upper limit of kd (in an acidic pH range condition)/kd (in a neutral pH range condition) value is not particularly limited, and may be any value such as 50, 100, or 200, as long as the molecule can be produced by the techniques of those skilled in the art.


When the antigen is a soluble antigen, the dissociation rate constant (kd) can be used as the value for antigen-binding activity and when the antigen is a membrane antigen, the apparent dissociation rate constant (kd) can be used. The dissociation rate constant (kd) and apparent dissociation rate constant (kd) can be determined by methods known to those skilled in the art, and Biacore (GE healthcare), flow cytometer, and such may be used. In the present invention, when the antigen-binding activity of an antigen-binding molecule is measured at different proton concentrations, i.e., pHs, conditions other than the proton concentration, i.e., pH, are preferably the same.


For example, an antigen-binding domain (or antigen-binding molecule) whose antigen-binding activity at a condition of high proton concentration or low pH, i.e., in an acidic pH range condition is lower than that at a condition of low proton concentration or high pH, i.e., in a neutral pH range condition, which is one embodiment provided by the present invention, can be obtained via screening of antigen-binding domains (or antigen-binding molecules), comprising the following steps (a) to (c):

  • (a) obtaining the antigen-binding activity of an antigen-binding domain (or antigen-binding molecule) in an acidic pH range condition;
  • (b) obtaining the antigen-binding activity of an antigen-binding domain (or antigen-binding molecule) in a neutral pH range condition; and
  • (c) selecting an antigen-binding domain (or antigen-binding molecule) whose antigen-binding activity in the acidic pH range condition is lower than that in the neutral pH range condition.


Alternatively, an antigen-binding domain (or antigen-binding molecule) whose antigen-binding activity at a condition of high proton concentration or low pH, i.e., in an acidic pH range condition, is lower than that at a condition of low proton concentration or high pH, i.e., in a neutral pH range condition, which is one embodiment provided by the present invention, can be obtained via screening of antigen-binding domains (or antigen-binding molecules), or a library thereof, comprising the following steps (a) to (c):

  • (a) contacting an antigen-binding domain (or antigen-binding molecule), or a library thereof, in a neutral pH range condition with an antigen;
  • (b) placing in an acidic pH range condition the antigen-binding domain (or antigen-binding molecule) bound to the antigen in step (a); and
  • (c) isolating the antigen-binding domain (or antigen-binding molecule) dissociated in step (b).


An antigen-binding domain (or antigen-binding molecule) whose antigen-binding activity at a condition of high proton concentration or low pH, i.e., in an acidic pH range condition is lower than that at a condition of low proton concentration or high pH, i.e., in a neutral pH range condition, which is another embodiment provided by the present invention, can be obtained via screening of antigen-binding domains (or antigen-binding molecules), or a library thereof, comprising the following steps (a) to (d):

  • (a) contacting in an acidic pH range an antigen with a library of antigen-binding domains (or antigen-binding molecules);
  • (b) selecting the antigen-binding domain (or antigen-binding molecule) which does not bind to the antigen in step (a);
  • (c) allowing the antigen-binding domain (or antigen-binding molecule) selected in step (b) to bind with the antigen in a neutral pH range; and
  • (d) isolating the antigen-binding domain (or antigen-binding molecule) bound to the antigen in step (c).


An antigen-binding domain (or antigen-binding molecule) whose antigen-binding activity at a condition of high proton concentration or low pH, i.e., in an acidic pH range condition, is lower than that at a condition of low proton concentration or high pH, i.e., in a neutral pH range condition, which is even another embodiment provided by the present invention, can be obtained by a screening method comprising the following steps (a) to (c):

  • (a) contacting in a neutral pH range condition a library of antigen-binding domains (or antigen-binding molecules) with a column immobilized with an antigen;
  • (b) eluting in an acidic pH range condition from the column the antigen-binding domain (or antigen-binding molecule) bound to the column in step (a); and
  • (c) isolating the antigen-binding domain (or antigen-binding molecule) eluted in step (b).


An antigen-binding domain (or antigen-binding molecule) whose antigen-binding activity at a condition of high proton concentration or low pH, i.e., in an acidic pH condition, range is lower than that at a condition of low proton concentration or high pH, i.e., in a neutral pH range condition, which is still another embodiment provided by the present invention, can be obtained by a screening method comprising the following steps (a) to (d):

  • (a) allowing, in an acidic pH range condition, a library of antigen-binding domains (or antigen-binding molecules) to pass a column immobilized with an antigen;
  • (b) collecting the antigen-binding domain (or antigen-binding molecule) eluted without binding to the column in step (a);
  • (c) allowing the antigen-binding domain (or antigen-binding molecule) collected in step (b) to bind with the antigen in a neutral pH range condition; and
  • (d) isolating the antigen-binding domain (or antigen-binding molecule) bound to the antigen in step (c).


An antigen-binding domain (or antigen-binding molecule) whose antigen-binding activity at a condition of high proton concentration or low pH, i.e., in an acidic pH range condition, is lower than that at a condition of low proton concentration or high pH, i.e., in a neutral pH range condition, which is yet another embodiment provided by the present invention, can be obtained by a screening method comprising the following steps (a) to (d):

  • (a) contacting an antigen with a library of antigen-binding domains (or antigen-binding molecules) in a neutral pH range condition;
  • (b) obtaining the antigen-binding domain (or antigen-binding molecule) bound to the antigen in step (a);
  • (c) placing in an acidic pH range condition the antigen-binding domain (or antigen-binding molecule) obtained in step (b); and
  • (d) isolating the antigen-binding domain (or antigen-binding molecule) whose antigen-binding activity in step (c) is weaker than the standard selected in step (b).


The above-described steps may be repeated twice or more times. Thus, the present invention provides antigen-binding domains (or antigen-binding molecules) whose antigen-binding activity in an acidic pH range condition is lower than that in a neutral pH range condition, which are obtained by a screening method that further comprises the steps of repeating twice or more times steps (a) to (c) or (a) to (d) in the above-described screening methods. The number of times that steps (a) to (c) or (a) to (d) is repeated is not particularly limited; however, the number is 10 or less in general.


In the screening methods of the present invention, the antigen-binding activity of an antigen-binding domain (or antigen-binding molecule) at a condition of high proton concentration or low pH, i.e., in an acidic pH range, is not particularly limited, as long as it is the antigen-binding activity at a pH of between 4.0 and 6.5, and includes the antigen-binding activity at a pH of between 4.5 and 6.6 as the preferred pH. The antigen-binding activity also includes that at a pH of between 5.0 and 6.5, and that at a pH of between 5.5 and 6.5 as another preferred pH. The antigen-binding activity also includes that at the pH in the early endosome in vivo as the more preferred pH, and specifically, that at pH 5.8. Meanwhile, the antigen-binding activity of an antigen-binding domain (or antigen-binding molecule) at a condition of low proton concentration or high pH, i.e., in a neutral pH range, is not particularly limited, as long as it is the antigen-binding activity at a pH of between 6.7 and 10, and includes the antigen-binding activity at a pH of between 6.7 and 9.5 as the preferred pH. The antigen-binding activity also includes that at a pH of between 7.0 and 9.5 and that at a pH of between 7.0 and 8.0 as another preferred pH. The antigen-binding activity also includes that at the pH of plasma in vivo as the more preferred pH, and specifically, that at pH 7.4.


The antigen-binding activity of an antigen-binding domain (or antigen-binding molecule) can be measured by methods known to those skilled in the art. Those skilled in the art can suitably determine conditions other than ionized calcium concentration. The antigen-binding activity of an antigen-binding domain (or antigen-binding molecule) can be assessed based on the dissociation constant (KD), apparent dissociation constant (1(D), dissociation rate constant (kd), apparent dissociation rate constant (kd), and such. These can be determined by methods known to those skilled in the art, for example, using Biacore (GE healthcare), Scatchard plot, or FACS.


In the present invention, the step of selecting an antigen-binding domain (or antigen-binding molecule) whose antigen-binding activity at a condition of low proton concentration or high pH, i.e., in a neutral pH range condition, is higher than that at a condition of high proton concentration or low pH, i.e., in an acidic pH range condition, is synonymous with the step of selecting an antigen-binding domain (or antigen-binding molecule) whose antigen-binding activity at a condition of high proton concentration or low pH, i.e., in an acidic pH range condition, is lower than that at a condition of low proton concentration or high pH, i.e., in a neutral pH range condition.


As long as the antigen-binding activity at a condition of low proton concentration or high pH, i.e., in a neutral pH range condition, is higher than that at a condition of high proton concentration or low pH, i.e., in an acidic pH range condition, the difference between the antigen-binding activity at a condition of low proton concentration or high pH, i.e., a neutral pH range condition, and that at a condition of high proton concentration or low pH, i.e., an acidic pH range condition, is not particularly limited; however, the antigen-binding activity at a condition of low proton concentration or high pH, i.e., in a neutral pH range condition, is preferably twice or more, more preferably 10 times or more, and still more preferably 40 times or more than that at a condition of high proton concentration or low pH, i.e., in an acidic pH range condition.


The antigen binding domain (or antigen-binding molecule) of the present invention that is screened by the screening methods described above may be any antigen-binding domain (or antigen-binding molecule), and for example, an above-mentioned antigen-binding domain (or antigen-binding molecule) may be screened. For example, antigen-binding domains (or antigen-binding molecules) having a native sequence may be screened, and antigen-binding domains (or antigen-binding molecules) in which their amino acid sequences have been substituted may also be screened.


The antigen-binding domain (or antigen-binding molecule) of the present invention to be screened by the above-described screening methods may be prepared in any manner. For example, preexisting antibodies, preexisting libraries (phage library, etc.), antibodies or libraries prepared from B cells of immunized animals or from hybridomas obtained by immunizing animals, antibodies or libraries (libraries with increased content of amino acids with a side chain pKa of 4.0-8.0 (for example, histidine and glutamic acid) or unnatural amino acids, libraries introduced with amino acids with a side chain pKa of 4.0-8.0 (for example, histidine and glutamic acid) or unnatural amino acid mutations at specific positions, etc.) obtained by introducing amino acids with a side chain pKa of 4.0-8.0 (for example, histidine and glutamic acid) or unnatural amino acid mutations into the above-described antibodies or libraries may be used.


Methods for obtaining an antigen-binding domain (or antigen-binding molecule) whose antigen-binding activity at a condition of low proton concentration or high pH, i.e., in a neutral pH range condition, is higher than that at a condition of high proton concentration or low pH, i.e., in an acidic pH range condition, from an antigen-binding domains (or antigen-binding molecules) prepared from hybridomas obtained by immunizing animals or from B cells of immunized animals preferably include, for example, the antigen-binding molecule or antibody in which at least one of the amino acids of the antigen-binding domain or antibody is substituted with an amino acid with a side chain pKa of 4.0-8.0 (for example, histidine and glutamic acid) or an unnatural amino acid mutation, or the antigen-binding domain or antibody inserted with an amino acid with a side chain pKa of 4.0-8.0 (for example, histidine and glutamic acid) or unnatural amino acid, such as those described in International Publication No. WO 2009/125825.


The sites of introducing mutations of amino acids with a side chain pKa of 4.0-8.0 (for example, histidine and glutamic acid) or unnatural amino acids are not particularly limited, and may be any position as long as the antigen-binding activity in an acidic pH range becomes weaker than that in a neutral pH range (the value of KD (in an acidic pH range)/KD (in a neutral pH range) or kd (in an acidic pH range)/kd (in a neutral pH range) is increased) as compared to before substitution or insertion. For example, when the antigen-binding molecule is an antibody, antibody variable region and CDRs are suitable. Those skilled in the art can appropriately determine the number of amino acids to be substituted with or the number of amino acids with a side chain pKa of 4.0-8.0 (for example, histidine and glutamic acid) or unnatural amino acids to be inserted. It is possible to substitute with a single amino acid having a side chain pKa of 4.0-8.0 (for example, histidine and glutamic acid) or a single unnatural amino acid; it is possible to insert a single amino acid having a side chain pKa of 4.0-8.0 (for example, histidine and glutamic acid) or a single unnatural amino acid; it is possible to substitute with two or more amino acids having a side chain pKa of 4.0-8.0 (for example, histidine and glutamic acid) or two or more unnatural amino acids; and it is possible to insert two or more amino acids having a side chain pKa of 4.0-8.0 (for example, histidine and glutamic acid) or two or more unnatural amino acids. Alternatively, other amino acids can be deleted, added, inserted, and/or substituted concomitantly, aside from the substitution into amino acids having a side chain pKa of 4.0-8.0 (for example, histidine and glutamic acid) or unnatural amino acids, or the insertion of amino acids having a side chain pKa of 4.0-8.0 (for example, histidine and glutamic acid) or unnatural amino acids. Substitution into or insertion of amino acids with a side chain pKa of 4.0-8.0 (for example, histidine and glutamic acid) or unnatural amino acids can performed randomly by methods such as histidine scanning, in which the alanine of alanine scanning known to those skilled in the art is replaced with histidine. Antigen-binding molecules exhibiting a greater value of KD (in an acidic pH range)/KD (in a neutral pH range) or kd (in an acidic pH range)/kd (in a neutral pH range) as compared to before the mutation can be selected from antigen-binding domains or antibodies introduced with random insertions or substitution mutations of amino acids with a side chain pKa of 4.0-8.0 (for example, histidine and glutamic acid) or unnatural amino acids.


Preferred examples of antigen-binding molecules containing the mutation into amino acids with a side chain pKa of 4.0-8.0 (for example, histidine and glutamic acid) or unnatural amino acids as described above and whose antigen-binding activity in an acidic pH range is lower than that in a neutral pH range include, antigen-binding molecules whose antigen-binding activity in the neutral pH range after the mutation into amino acids with a side chain pKa of 4.0-8.0 (for example, histidine and glutamic acid) or unnatural amino acids is comparable to that before the mutation into amino acids with a side chain pKa of 4.0-8.0 (for example, histidine and glutamic acid) or unnatural amino acids. Herein, “an antigen-binding molecule after the mutation with amino acids having a side chain pKa of 4.0-8.0 (for example, histidine and glutamic acid) or unnatural amino acids has an antigen-binding activity comparable to that before the mutation with amino acids having a side chain pKa of 4.0-8.0 (for example, histidine and glutamic acid) or unnatural amino acids” means that, when taking the antigen-binding activity of an antigen-binding molecule before the mutation with amino acids having a side chain pKa of 4.0-8.0 (for example, histidine and glutamic acid) or unnatural amino acids as 100%, the antigen-binding activity of an antigen-binding molecule after the mutation with amino acids having a side chain pKa of 4.0-8.0 (for example, histidine and glutamic acid) or unnatural amino acids is at least 10% or more, preferably 50% or more, more preferably 80% or more, and still more preferably 90% or more. The antigen-binding activity after the mutation of amino acids with a side chain pKa of 4.0-8.0 (for example, histidine and glutamic acid) or unnatural amino acids at pH 7.4 may be higher than that before the mutation of amino acids with a side chain pKa of 4.0-8.0 (for example, histidine and glutamic acid) or unnatural amino acids at pH 7.4. If the antigen-binding activity of an antigen-binding molecule is decreased due to insertion of or substitution into amino acids with a side chain pKa of 4.0-8.0 (for example, histidine and glutamic acid) or unnatural amino acids, the antigen-binding activity can be made to be comparable to that before the insertion of or substitution into amino acids with a side chain pKa of 4.0-8.0 (for example, histidine and glutamic acid) or unnatural amino acids, by introducing a substitution, deletion, addition, and/or insertion of one or more amino acids of the antigen-binding molecule. The present invention also includes antigen-binding molecules whose binding activity has been adjusted to be comparable by substitution, deletion, addition, and/or insertion of one or more amino acids after substitution or insertion of amino acids with a side chain pKa of 4.0-8.0 (for example, histidine and glutamic acid) or unnatural amino acids.


Amino Acids that Alter the Antigen-Binding Activity of Antigen-Binding Domain Depending on the Proton Concentration Conditions


Antigen-binding domains (or antigen-binding molecules) of the present invention to be screened by the above-described screening methods may be prepared in any manner. For example, when ion concentration condition is proton concentration condition or pH condition, preexisting antibodies, preexisting libraries (phage library, etc.), antibodies or libraries prepared from B cells of immunized animals or from hybridomas obtained by immunizing animals, antibodies or libraries (libraries with increased content of amino acids with a side chain pKa of 4.0-8.0 (for example, histidine and glutamic acid) or unnatural amino acids, libraries introduced with mutations of amino acids with a side chain pKa of 4.0-8.0 (for example, histidine and glutamic acid) or unnatural amino acids at specific positions, etc.) obtained by introducing mutations of amino acids with a side chain pKa of 4.0-8.0 (for example, histidine and glutamic acid) or unnatural amino acids into the above-described antibodies or libraries may be used.


In one embodiment of the present invention, a library containing multiple antigen-binding molecules of the present invention whose sequences are different from one another can also be constructed by combining heavy chain variable regions, produced as a randomized variable region sequence library, with light chain variable regions introduced with “at least one amino acid residue that changes the antigen-binding activity of an antigen-binding molecule depending on the proton concentration condition”.


Such amino acid residues include, but are not limited to, for example, amino acid residues contained in the light chain CDR1. The amino acid residues also include, but are not limited to, for example, amino acid residues contained in the light chain CDR2. The amino acid residues also include, but are not limited to, for example, amino acid residues contained in the light chain CDR3.


The above-described amino acid residues contained in the light chain CDR1 include, but are not limited to, for example, amino acid residues of positions 24, 27, 28, 31, 32, and/or 34 according to Kabat numbering in the CDR1 of light chain variable region. Meanwhile, the amino acid residues contained in the light chain CDR2 include, but are not limited to, for example, amino acid residues of positions 50, 51, 52, 53, 54, 55, and/or 56 according to Kabat numbering in the CDR2 of light chain variable region. Furthermore, the amino acid residues in the light chain CDR3 include, but are not limited to, for example, amino acid residues of positions 89, 90, 91, 92, 93, 94, and/or 95A according to Kabat numbering in the CDR3 of light chain variable region. Moreover, the amino acid residues can be contained alone or can be contained in combination of two or more amino acids as long as they allow the change in the antigen-binding activity of an antigen-binding molecule depending on the proton concentration condition.


Even when the heavy chain variable region produced as a randomized variable region sequence library is combined with the above-described light chain variable region introduced with “at least one amino acid residue that changes the antigen-binding activity of an antigen-binding molecule depending on the proton concentration condition”, it is possible to design so that the flexible residues are contained in the sequence of the light chain variable region in the same manner as described above. The number and position of the flexible residues are not particularly limited to a specific embodiment, as long as the antigen-binding activity of an antigen-binding molecule of the present invention changes depending on the proton concentration condition. Specifically, the CDR and/or FR sequences of heavy chain and/or light chain can contain one or more flexible residues. For example, flexible residues to be introduced into the sequences of the light chain variable regions include, but are not limited to, for example, the amino acid residues listed in Tables 3 and 4. Meanwhile, amino acid sequences of light chain variable regions other than the flexible residues and amino acid residues that change the antigen-binding activity of an antigen-binding molecule depending on the proton concentration condition suitably include, but are not limited to, germ line sequences such as Vk1 (SEQ ID NO: 3), Vk2 (SEQ ID NO: 4), Vk3 (SEQ ID NO: 5), and Vk4 (SEQ ID NO: 6).










TABLE 3





Position
Amino acid















CDR1
















28
S: 100%










29
I: 100%


30
N: 25%
S: 25%
R: 25%
H: 25%


31
S: 100%


32
H: 100%


33
L: 100%


34
A: 50%
N: 50%







CDR2
















50
H: 100%



or
A: 25%
D: 25%
G: 25%
K: 25%


51
A: 100%




A: 100%


52
S: 100%




S: 100%


53
K: 33.3%
N: 33.3%
S: 33.3%


H: 100%


54
L: 100%




L: 100%


55
Q: 100%




Q: 100%


56
S: 100%




S: 100%







CDR3
















90
Q: 100%



or
Q: 100%





91
H: 100%




8: 33.3%
R: 33.3%
Y: 33.3%


92
G: 25%
N: 25%
S: 25%
Y:25%

H: 100%


93
H: 33.3%
N: 33.3%
S: 33.3%


H: 33.3%
N: 33.3%
S: 33.3%


94
S: 50%
Y: 50%



S: 50%
Y: 50%


95
P: 100%




P: 100%


96
L: 50%
Y: 50%



L: 50%
Y: 50%





(Position indicates Kabat numbering)






When position 92 as indicated by Kabat numbering is Asn (N), Ser (S) at position 94 may be excluded.











TABLE 4





CDR
Position
Amino acid





















CDR1
28
S: 100%







29
I: 100%



30
H: 30%
N: 10%
S: 50%
R: 10%



31
N: 35%
S: 65%



32
H: 40%
N: 20%
Y: 40%



33
L: 100%



34
A: 70%
N: 30%


CDR2
50
A: 25%
D: 15%
G: 25%
H: 30%
K: 5%



51
A: 100%



52
S: 100%



S3
H: 30%
K: 10%
N: 15%
S: 45%



54
L: 100%



55
Q: 100%



56
S: 100%


CDR3
90
Q: 100%



91
H: 30%
S: 15%
R: 10%
Y: 45%



92
G: 20%
H: 30%
N: 20%
S: 15%
Y: 15%



93
H: 30%
N: 25%
S: 45%



94
S: 50%
Y: 50%



95
P: 100%



96
L: 50%
Y: 50%





(Position indicates Kabat numbering)






When position 92 as indicated by Kabat numbering is Asn (N), Ser (S) at position 94 may be excluded.


Any amino acid residue may be suitably used as the above-described amino acid residues that change the antigen-binding activity of an antigen-binding molecule depending on the proton concentration condition. Specifically, such amino acid residues include amino acids with a side chain pKa of 4.0-8.0. Such electron-releasing amino acids preferably include, for example, naturally occurring amino acids such as histidine and glutamic acid, as well as unnatural amino acids such as histidine analogs (US2009/0035836), m-NO2-Tyr (pKa 7.45), 3,5-Br2-Tyr (pKa 7.21), and 3,5-12-Tyr (pKa 7.38) (Bioorg. Med. Chem. (2003) 11 (17), 3761-3768). Particularly preferred amino acid residues include, for example, amino acids with a side chain pKa of 6.0-7.0. Such electron-releasing amino acid residues preferably include, for example, histidine.


Known methods such as site-directed mutagenesis (Kunkel et al. (Proc. Natl. Acad. Sci. USA (1985) 82, 488-492)) and Overlap extension PCR can be appropriately employed to alter the amino acids of antigen-binding domains. Furthermore, various known methods can also be used as an amino acid alteration method for substituting amino acids by those other than natural amino acids (Annu Rev. Biophys. Biomol. Struct. (2006) 35: 225-249; Proc. Natl. Acad. Sci. U.S.A. (2003) 100(11): 6353-6357). For example, a cell-free translation system (Clover Direct (Protein Express)) containing tRNAs in which amber suppressor tRNA, which is complementary to UAG codon (amber codon) that is a stop codon, is linked with an unnatural amino acid may be suitably used.


The preferred heavy chain variable region that is used in combination includes, for example, randomized variable region libraries. Known methods are appropriately combined as a method for producing a randomized variable region library. In a non-limiting embodiment of the present invention, an immune library constructed based on antibody genes derived from animals immunized with specific antigens, patients with infection or persons with an elevated antibody titer in blood as a result of vaccination, cancer patients, or lymphocytes of auto immune diseases may be suitably used as a randomized variable region library.


In another non-limiting embodiment of the present invention, in the same manner as described above, a synthetic library in which the CDR sequences of V genes from genomic DNA or functional reconstructed V genes are replaced with a set of synthetic oligonucleotides containing the sequences encoding codon sets of an appropriate length can also be suitably used as a randomized variable region library. In this case, the CDR3 sequence alone may be replaced because variety in the gene sequence of heavy chain CDR3 is observed. The basis for giving rise to amino acid variations in the variable region of an antigen-binding molecule is to generate variations of amino acid residues of surface-exposed positions of the antigen-binding molecule. The surface-exposed position refers to a position where an amino acid is exposed on the surface and/or contacted with an antigen based on the conformation, structural ensemble, and/or modeled structure of an antigen-binding molecule, and in general, such positions are the CDRs. The surface-exposed positions are preferably determined using the coordinates derived from a three-dimensional model of the antigen-binding molecule using computer programs such as Insightll program (Accelrys). The surface-exposed positions can be determined using algorithms known in the art (for example, Lee and Richards (J. Mol. Biol. (1971) 55, 379-400); Connolly (J. Appl. Cryst. (1983) 16, 548-558)). The surface-exposed positions can be determined based on the information on the three dimensional structure of antibodies using software suitable for protein modeling. Software which is suitably used for this purpose includes the SYBYL biopolymer module software (Tripos Associates). When the algorithm requires the input size parameter from the user, the “size” of probe for use in computation is generally or preferably set at about 1.4 angstrom or less in radius. Furthermore, a method for determining surface-exposed region and area using personal computer software is described by Pacios (Comput. Chem. (1994) 18 (4), 377-386; and J. Mol. Model. (1995) 1, 46-53).


In still another non-limiting embodiment of the present invention, a naive library constructed from antibody genes derived from lymphocytes of healthy persons and consisting of naive sequences, which are unbiased repertoire of antibody sequences, can also be particularly suitably used as a randomized variable region library (Gejima et al. (Human Antibodies (2002) 11, 121-129); and Cardoso et al. (Scand. J. Immunol. (2000) 51, 337-344)).


Fc Region

An Fc region contains the amino acid sequence derived from the heavy chain constant region of an antibody. An Fc region is a portion of the heavy chain constant region of an antibody, starting from the N terminal end of the hinge region, which corresponds to the papain cleavage site at an amino acid around position 216 according to the EU numbering system, and contains the hinge, CH2, and CH3 domains. While the Fc region may be obtained from human IgG1, it is not limited to a particular subclass of IgG. As described later, a favorable example of the Fc region is an Fc region that has an FcRn-binding activity in an acidic pH range. Furthermore, a favorable example of the Fc region is an Fc region that has an Fcγ receptor-binding activity as described later. A non-limiting embodiment of such an Fc region is, for example, the Fc region of human IgG1 (SEQ ID NO: 9), IgG2 (SEQ ID NO: 10), IgG3 (SEQ ID NO: 11), or IgG4 (SEQ ID NO: 12). A number of allotype sequences of human IgG1, human IgG2, human IgG3, and human IgG4 constant regions due to gene polymorphisms are described in “Sequences of proteins of immunological interest”, NIH Publication No. 91-3242. Any of such sequences may be used in the present invention. In particular, for the human IgG1 sequence, the amino acid sequence at positions 356 to 358 as indicated by EU numbering may be DEL or EEM. Furthermore, an Fc region does not have to be derived from the above-described human IgG constant region as long as it has a domain that binds to FcγR and/or FcRn.


FcRn

Unlike Fcγ receptor belonging to the immunoglobulin superfamily, human FcRn is structurally similar to polypeptides of major histocompatibility complex (MHC) class I, exhibiting 22% to 29% sequence identity to class I MHC molecules (Ghetie el al., Immunol. Today (1997) 18 (12): 592-598). FcRn is expressed as a heterodimer consisting of soluble β or light chain (β2 microglobulin) complexed with transmembrane a or heavy chain. Like MHC, FcRn a chain comprises three extracellular domains (α1, α2, and α3) and its short cytoplasmic domain anchors the protein onto the cell surface. α1 and α2 domains interact with the FcRn-binding domain of the antibody Fc region (Raghavan et al., Immunity (1994) 1: 303-315).


FcRn is expressed in maternal placenta and york sac of mammals, and is involved in mother-to-fetus IgG transfer. In addition, in neonatal small intestine of rodents, where FcRn is expressed, FcRn is involved in transfer of maternal IgG across brush border epithelium from ingested colostrum or milk. FcRn is expressed in a variety of other tissues and endothelial cell systems of various species. FcRn is also expressed in adult human endothelia, muscular blood vessels, and hepatic sinusoidal capillaries. FcRn is believed to play a role in maintaining the plasma IgG concentration by mediating recycling of IgG to serum upon binding to IgG. Typically, binding of FcRn to IgG molecules is strictly pH dependent. The optimal binding is observed in an acidic pH range below 7.0.


Human FcRn whose precursor is a polypeptide having the signal sequence of SEQ ID


NO: 13 (the polypeptide with the signal sequence is shown in SEQ ID NO: 14) forms a complex with human β2-microglobulin in vivo. Soluble human FcRn complexed with β2-microglobulin is produced by using conventional recombinant expression techniques. Fc regions of the present invention can be assessed for their binding activity to such a soluble human FcRn complexed with β2-microglobulin. In the present invention, unless otherwise specified, human FcRn refers to a form capable of binding to an Fc region of the present invention. Examples include a complex between human FcRn and human β2-microglobulin.


Binding Activity of the Fc Region to FcRn, in Particular, Human FcRn

The binding activity of an Fc region of the present invention to FcRn, human FcRn in particular, can be measured by methods known to those skilled in the art, as described in the section “Binding Activity” above. Those skilled in the art can appropriately determine the conditions other than pH. The antigen-binding activity and human FcRn-binding activity of an antigen-binding molecule can be assessed based on the dissociation constant (KD), apparent dissociation constant (KD), dissociation rate (kd), apparent dissociation rate (kd), and such. These can be measured by methods known to those skilled in the art. For example, Biacore (GE healthcare), Scatchard plot, or flow cytometer may be used.


When the human FcRn-binding activity of an Fc region of the present invention is measured, conditions other than the pH are not particularly limited, and can be appropriately selected by those skilled in the art. Measurements can be carried out, for example, at 37° C. using MES buffer, as described in International Publication No. WO 2009/125825. Alternatively, the human FcRn-binding activity of an Fc region of the present invention can be measured by methods known to those skilled in the art, and may be measured by using, for example, Biacore (GE Healthcare) or such. The binding activity of an Fc region of the present invention to human FcRn can be assessed by pouring, as an analyte, human FcRn, an Fc region, or an antigen-binding molecule of the present invention containing the Fc region into a chip immobilized with an Fc region, an antigen-binding molecule of the present invention containing the Fc region, or human FcRn.


A neutral pH range as the condition where the Fc region contained in an antigen-binding molecule of the present invention has the FcRn-binding activity means pH 6.7 to pH 10.0 in general. Preferably, the neutral pH range is a range indicated with arbitrary pH values between pH 7.0 and pH 8.0, and is preferably selected from pH 7.0, 7.1, 7.2, 7.3, 7.4, 7.5, 7.6, 7.7, 7.8, 7.9, and 8.0, and is particularly preferably pH 7.4 that is close to the pH of plasma (blood) in vivo. When the binding affinity between the human FcRn-binding domain and human FcRn at pH 7.4 is too low to assess, pH 7.0 may be used instead of pH 7.4. Herein, an acidic pH range as the condition where the Fc region contained in an antigen-binding molecule of the present invention has the FcRn-binding activity means pH 4.0 to pH 6.5 in general. Preferably, the acidic pH range means pH 5.5 to pH 6.5, particularly preferably pH 5.8 to pH 6.0 which is close to the pH in the early endosome in vivo. Regarding the temperature used as the measurement condition, the binding affinity between the human FcRn-binding domain and human FcRn may be assessed at any temperature between 10° C. and 50° C. Preferably, the binding affinity between the human FcRn-binding domain and human FcRn can be determined at 15° C. to 40° C. More preferably, the binding affinity between the human FcRn-binding domain and human FcRn can be determined in the same manner at an arbitrary temperature between 20° C. and 35° C., such as any one temperature of 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, and 35° C. In an embodiment of the present invention, the temperature includes, but is not limited to, for example, 25° C.


According to the Journal of Immunology (2009) 182, 7663-7671, the human FcRn-binding activity of native human IgG1 in an acidic pH range (pH 6.0) is 1.7 μM (1(D), and the activity is almost undetectable in a neutral pH range. Thus, in a preferred embodiment, antigen-binding molecules comprising an Fc region of which human FcRn-binding activity in an acidic pH range is 20 μM (KD) or stronger may be screened. In a more preferred embodiment, the antigen-binding molecules comprising an Fc region of which human FcRn-binding activity in an acidic pH range is 2.0 μM (KD) or stronger may be screened. In a still more preferred embodiment, the antigen-binding molecules comprising an Fc region of which human FcRn-binding activity in an acidic pH range is 0.5 μM (KD) or stronger may be screened. The above-mentioned KD values are determined by the method described in the Journal of Immunology (2009) 182: 7663-7671 (by immobilizing the antigen-binding molecule onto a chip and loading human FcRn as an analyte).


In the present invention, preferred Fc regions have an FcRn-binding activity under an acidic pH range condition. When an Fc region originally has an FcRn-binding activity under an acidic pH range condition, the domain can be used as it is. When the domain has a weak or no FcRn-binding activity under an acidic pH range condition, an Fc region having a desired FcRn-binding activity can be obtained by altering amino acids of an antigen-binding molecule. Fc regions having a desired or enhanced FcRn-binding activity under an acidic pH range condition can also be suitably obtained by altering the amino acids of an Fc region. Amino acid alterations of an Fc region that result in such a desired binding activity can be found by comparing the FcRn-binding activity under an acidic pH range condition before and after amino acid alteration. Those skilled in the art can appropriately alter the amino acids using known techniques similar to the aforementioned techniques used to alter the Fcγ-receptor-binding activity.


Fc regions comprised in the antigen-binding molecules of the present invention, which have an FcRn-binding activity under an acidic pH range condition, can be obtained by any method. Specifically, FcRn-binding domains having an FcRn-binding activity or an enhanced FcRn-binding activity under an acidic pH range condition can be obtained by altering the amino acids of an IgG-type human immunoglobulin used as a starting Fc region. Preferred Fc regions of an IgG-type immunoglobulin for alteration include, for example, those of human IgGs (IgG1, IgG2, IgG3, and IgG4, and variants thereof). As long as the Fc region has an FcRn-binding activity under an acidic pH range condition or can increase the human FcRn-binding activity under an acidic pH range condition, amino acids at any position may be altered into other amino acids. When the antigen-binding molecule contains the Fc region of human IgG1 as the Fc region, it is preferable that the resulting Fc region contains an alteration that results in the effect of enhancing FcRn binding under an acidic pH range condition as compared to the binding activity of the starting human IgG1 Fc region. Amino acids that allow such alteration include, for example, at least one or more amino acids selected from the group of positions 252, 254, 256, 309, 311, 315, 433, and 434 according to EU numbering, and their combination amino acids of at least one or more amino acids selected from the group of positions 253, 310, 435, and 426 as described in WO 1997/034631. Favorable examples include at least one or more amino acids selected from the group of positions 238, 252, 253, 254, 255, 256, 265, 272, 286, 288, 303, 305, 307, 309, 311, 312, 317, 340, 356, 360, 362, 376, 378, 380, 382, 386, 388, 400, 413, 415, 424, 433, 434, 435, 436, 439, and 447 as indicated by EU numbering as described in WO 2000/042072. Similarly, favorable examples of amino acids that allow such alteration include, at least one or more amino acids selected from the group of positions 251, 252, 254, 255, 256, 308, 309, 311, 312, 385, 386, 387, 389, 428, 433, 434, and 436 according to EU numbering as described in WO 2002/060919. Furthermore, amino acids that allow such alteration include, for example, amino acids of positions 250, 314, and 428 according to EU numbering as described in WO2004/092219. In addition, favorable examples of amino acids that allow such alteration include at least one or more amino acids selected from the group of positions 238, 244, 245, 249, 252, 256, 257, 258, 260, 262, 270, 272, 279, 283, 285, 286, 288, 293, 307, 311, 312, 316, 317, 318, 332, 339, 341, 343, 375, 376, 377, 378, 380, 382, 423, 427, 430, 431, 434, 436, 438, 440, and 442 as described in WO 2006/020114. Furthermore, favorable examples of amino acids that allow such alteration include amino acids of positions 251, 252, 307, 308, 378, 428, 430, 434, and/or 436 according to EU numbering as described in WO 2010/045193. Alteration of these amino acids enhances FcRn binding of the Fc region of an IgG-type immunoglobulin under an acidic pH range condition.


When the Fc region of human IgG1 is comprised as the Fc region, a non-limiting embodiment of the alteration that results in the effect of enhancing FcRn binding under an acidic pH range condition as compared to the binding activity of the starting Fc region of human IgG1 includes at least one or more amino acid alterations selected from the group consisting of:


Arg or Leu for the amino acid at position 251;


Phe, Ser, Thr, or Tyr for the amino acid at position 252;


Ser or Thr for the amino acid at position 254;


Arg, Gly, Ile, or Leu for the amino acid at position 255;


Ala, Arg, Asn, Asp, Gln, Glu, or Thr for the amino acid at position 256;


Ile or Thr for the amino acid at position 308;


Pro for the amino acid at position 309;


Glu, Leu, or Ser for the amino acid at position 311;


Ala or Asp for the amino acid at position 312;


Ala or Leu for the amino acid at position 314;


Ala, Arg, Asp, Gly, His, Lys, Ser, or Thr for the amino acid at position 385;


Arg, Asp, Ile, Lys, Met, Pro, Ser, or Thr for the amino acid at position 386;


Ala, Arg, His, Pro, Ser, or Thr for the amino acid at position 387;


Asn, Pro, or Ser for the amino acid at position 389;


Leu, Met, Phe, Ser, or Thr for the amino acid at position 428;


Arg, Gln, His, Ile, Lys, Pro, or Ser for the amino acid at position 433;


His, Phe, or Tyr for the amino acid at position 434; and


Arg, Asn, His, Lys, Met, or Thr for the amino acid at position 436, as indicated by EU numbering. Meanwhile, the number of amino acids to be altered is not particularly limited; and amino acid may be altered at only one site or amino acids may be altered at two or more sites.


When the Fc region of human IgG1 is comprised as the Fc region, a non-limiting embodiment of the alteration that results in the effect of enhancing FcRn binding in an acidic pH range condition as compared to the binding activity of the starting Fc region of human IgG1 may be alterations including Ile for the amino acid at position 308, Pro for the amino acid at position 309, and/or Glu for the amino acid at position 311 according to EU numbering. Another non-limiting embodiment of this alteration may include Thr for the amino acid at position 308, Pro for the amino acid at position 309, Leu for the amino acid at position 311, Ala for the amino acid at position 312, and/or Ala for the amino acid at position 314. Furthermore, another non-limiting embodiment of this alteration may include Ile or Thr for the amino acid at position 308, Pro for the amino acid at position 309, Glu, Leu, or Ser for the amino acid at position 311, Ala for the amino acid at position 312, and/or Ala or Leu for the amino acid at position 314. Another non-limiting embodiment of this alteration may include Thr for the amino acid at position 308, Pro for the amino acid at position 309, Ser for the amino acid at position 311, Asp for the amino acid at position 312, and/or Leu for the amino acid at position 314.


When the Fc region of human IgG1 is comprised as the Fc region, a non-limiting embodiment of the alteration that results in the effect of enhancing FcRn binding under an acidic pH range condition as compared to the binding activity of the starting Fc region of human IgG1 may be alterations including Leu for the amino acid at position 251, Tyr for the amino acid at position 252, Ser or Thr for the amino acid at position 254, Arg for the amino acid at position 255, and/or Glu for the amino acid at position 256 according to EU numbering.


When the Fc region of human IgG1 is comprised as the Fc region, a non-limiting embodiment of the alteration that results in the effect of enhancing FcRn binding under an acidic pH range condition as compared to the binding activity of the starting Fc region of human IgG1 may be at least one or more alterations selected from the group of alterations including Leu, Met, Phe, Ser, or Thr for the amino acid at position 428, Arg, Gln, His, Ile, Lys, Pro, or Ser for the amino acid at position 433, His, Phe, or Tyr for the amino acid at position 434, and/or Arg, Asn, His, Lys, Met, or Thr for the amino acid at position 436 according to EU numbering. Another non-limiting embodiment of this alteration may include His or Met for the amino acid at position 428, and/or His or Met for the amino acid at position 434.


When the Fc region of human IgG1 is comprised as the Fc region, a non-limiting embodiment of the alteration that results in the effect of enhancing FcRn binding under an acidic pH range condition as compared to the binding activity of the starting Fc region of human IgG1 may be alterations including Arg for the amino acid at position 385, Thr for the amino acid at position 386, Arg for the amino acid at position 387, and/or Pro for the amino acid at position 389 according to EU numbering. Another non-limiting embodiment of this alteration may include Asp for the amino acid at position 385, Pro for the amino acid at position 386, and/or Ser for the amino acid at position 389.


Furthermore, when the Fc region of human IgG1 is comprised as the Fc region, a non-limiting embodiment of the alteration that results in the effect of enhancing FcRn binding under an acidic pH range condition as compared to the binding activity of the starting Fc region of human IgG1 include at least one or more amino acid alterations selected from the group consisting of:


Gln or Glu for the amino acid at position 250; and


Leu or Phe for the amino acid at position 428 according to EU numbering. The number of amino acids to be altered is not particularly limited; and amino acid may be altered at only one site or amino acids may be altered at two sites.


When the Fc region of human IgG1 is comprised as the Fc region, a non-limiting embodiment of the alteration that results in the effect of enhancing FcRn binding under an acidic pH range condition as compared to the binding activity of the starting Fc region of human IgG1 may be alterations including Gln for the amino acid at position 250, and/or Leu or Phe for the amino acid at position 428 according to EU numbering. Another non-limiting embodiment of this alteration may include Glu for the amino acid at position 250, and/or Leu or Phe for the amino acid at position 428.


When the Fc region of human IgG1 is comprised as the Fc region, a non-limiting embodiment of the alteration that results in the effect of enhancing FcRn binding under an acidic pH range condition as compared to the binding activity of the starting Fc region of human IgG1 include at least two or more amino acid alterations selected from the group consisting of:


Asp or Glu for the amino acid at position 251;


Tyr for the amino acid at position 252;


Gln for the amino acid at position 307;


Pro for the amino acid at position 308;


Val for the amino acid at position 378;


Ala for the amino acid at position 380;


Leu for the amino acid at position 428;


Ala or Lys for the amino acid at position 430;


Ala, His, Ser, or Tyr for the amino acid at position 434; and


Ile for the amino acid at position 436, as indicated by EU numbering. The number of amino acids to be altered is not particularly limited; and amino acid may be altered at only two sites or amino acids may be altered at three or more sites.


When the Fc region of human IgG1 is comprised as the Fc region, a non-limiting embodiment of the alteration that results in the effect of enhancing FcRn binding under an acidic pH range condition as compared to the binding activity of the starting Fc region of human IgG1 may be alterations including Gln for the amino acid at position 307, and Ala or Ser for the amino acid at position 434 according to EU numbering. Another non-limiting embodiment of this alteration may include Pro for the amino acid at position 308, and Ala for the amino acid at position 434. Furthermore, another non-limiting embodiment of this alteration may include Tyr for the amino acid at position 252, and Ala for the amino acid at position 434. A different non-limiting embodiment of this alteration may include Val for the amino acid at position 378, and Ala for the amino acid at position 434. Another different non-limiting embodiment of this alteration may include Leu for the amino acid at position 428, and Ala for the amino acid at position 434. Another different non-limiting embodiment of this alteration may include Ala for the amino acid at position 434, and Ile for the amino acid at position 436. Furthermore, another non-limiting embodiment of this alteration may include Pro for the amino acid at position 308, and Tyr for the amino acid at position 434. In addition, another non-limiting embodiment of this alteration may include Gln for the amino acid at position 307, and Ile for the amino acid at position 436.


When the Fc region of human IgG1 is comprised as the Fc region, a non-limiting embodiment of the alteration that results in the effect of enhancing FcRn binding under an acidic pH range condition as compared to the binding activity of the starting Fc region of human IgG1 may be alterations including any one of Gln for the amino acid at position 307, Ala for the amino acid at position 380, and Ser for the amino acid at position 434 according to EU numbering. Another non-limiting embodiment of this alteration may include Gln for the amino acid at position 307, Ala for the amino acid at position 380, and Ala for the amino acid at position 434. Furthermore, another non-limiting embodiment of this alteration may include Tyr for the amino acid at position 252, Pro for the amino acid at position 308, and Tyr for the amino acid at position 434. A different non-limiting embodiment of this alteration may include Asp for the amino acid at position 251, Gln for the amino acid at position 307, and His for the amino acid at position 434.


When the Fc region of human IgG1 is comprised as the Fc region, a non-limiting embodiment of the alteration that results in the effect of enhancing FcRn binding under an acidic pH range condition as compared to the binding activity of the starting Fc region of human IgG1 include alteration of at least two or more amino acids selected from the group consisting of:


Leu for the amino acid at position 238;


Leu for the amino acid at position 244;


Arg for the amino acid at position 245;


Pro for the amino acid at position 249;


Tyr for the amino acid at position 252;


Pro for the amino acid at position 256;


Ala, Ile, Met, Asn, Ser, or Val for the amino acid at position 257;


Asp for the amino acid at position 258;


Ser for the amino acid at position 260;


Leu for the amino acid at position 262;


Lys for the amino acid at position 270;


Leu or Arg for the amino acid at position 272;


Ala, Asp, Gly, His, Met, Asn, Gln, Arg, Ser, Thr, Trp, or Tyr for the amino acid at position 279;


Ala, Asp, Phe, Gly, His, Ile, Lys, Leu, Asn, Pro, Gln, Arg, Ser, Thr, Trp, or Tyr for the amino acid


at position 283;


Asn for the amino acid at position 285;


Phe for the amino acid at position 286;


Asn or Pro for the amino acid at position 288;


Val for the amino acid at position 293;


Ala, Glu, or Met for the amino acid at position 307;


Ala, Ile, Lys, Leu, Met, Val, or Trp for the amino acid at position 311;


Pro for the amino acid at position 312;


Lys for the amino acid at position 316;


Pro for the amino acid at position 317;


Asn or Thr for the amino acid at position 318;


Phe, His, Lys, Leu, Met, Arg, Ser, or Trp for the amino acid at position 332;


Asn, Thr, or Trp for the amino acid at position 339;


Pro for the amino acid at position 341;


Glu, His, Lys, Gln, Arg, Thr, or Tyr for the amino acid at position 343;


Arg for the amino acid at position 375;


Gly, Ile, Met, Pro, Thr, or Val for the amino acid at position 376;


Lys for the amino acid at position 377;


Asp or Asn for the amino acid at position 378;


Asn, Ser, or Thr for the amino acid at position 380;


Phe, His, Ile, Lys, Leu, Met, Asn, Gln, Arg, Ser, Thr, Val, Trp, or Tyr for the amino acid at position 382;


Asn for the amino acid at position 423;


Asn for the amino acid at position 427;


Ala, Phe, Gly, His, Ile, Lys, Leu, Met, Asn, Gln, Arg, Ser, Thr, Val, or Tyr for the amino acid at position 430;


His or Asn for the amino acid at position 431;


Phe, Gly, His, Trp, or Tyr for the amino acid at position 434;


Ile, Leu, or Thr for the amino acid at position 436;


Lys, Leu, Thr, or Trp for the amino acid at position 438;


Lys for the amino acid at position 440; and


Lys for the amino acid at position 442 according to EU numbering. The number of amino acids to be altered is not particularly limited and amino acid at only two sites may be altered and amino acids at three or more sites may be altered.


When the Fc region of human IgG1 is comprised as the Fc region, a non-limiting embodiment of the alteration that results in the effect of enhancing FcRn binding under an acidic pH range condition as compared to the binding activity of the starting Fc region of human IgG1 may be alterations including Ile for the amino acid at position 257, and Ile for the amino acid at position 311 according to EU numbering. Another non-limiting embodiment of this alteration may include Ile for the amino acid at position 257, and His for the amino acid at position 434. Another non-limiting embodiment of this alteration may include Val for the amino acid at position 376, and His for the amino acid at position 434.


Furthermore, in another non-limiting embodiment, one may screen for antigen-binding molecules comprising an Fc region with the characteristic of having a human FcRn-binding activity in the neutral pH range instead of the above-described characteristic of having a human FcRn-binding activity in the acidic pH range. In a preferred embodiment, one may screen for antigen-binding molecules comprising an Fc region whose human FcRn-binding activity in the neutral pH range is 40 μM (KD) or stronger. In a more preferred embodiment, one may screen for antigen-binding molecules comprising an Fc region whose human FcRn-binding activity in the neutral pH range is 15 μM (KD) or stronger.


Furthermore, in another non-limiting embodiment, one may screen for antigen-binding molecules comprising an Fc region with the characteristic of having a human FcRn-binding activity in the neutral pH range in addition to the above-described characteristic of having a human FcRn-binding activity in the acidic pH range. In a preferred embodiment, one may screen for antigen-binding molecules comprising an Fc region whose human FcRn-binding activity in the neutral pH range is 40 μM (KD) or stronger. In a more preferred embodiment, one may screen for antigen-binding molecules comprising an Fc region whose human FcRn-binding activity in the neutral pH range is 15 μM (KD) or stronger.


In the present invention, preferred Fc regions have a human FcRn-binding activity in the acidic pH range and/or neutral pH range. When an Fc region originally has a human FcRn-binding activity in the acidic pH range and/or neutral pH range, it can be used as it is. When an Fc region has a weak or no human FcRn-binding activity in the acidic pH range and/or neutral pH range, antigen-binding molecules comprising an Fc region having a desired human FcRn-binding activity can be obtained by altering amino acids of the Fc region comprised in the antigen-binding molecules. Fc regions having a desired human FcRn-binding activity in the acidic pH range and/or neutral pH range can also be suitably obtained by altering amino acids of a human Fc region. Alternatively, antigen-binding molecules comprising an Fc region having a desired human FcRn-binding activity can be obtained by altering amino acids of an Fc region that originally has a human FcRn-binding activity in the acidic pH range and/or neutral pH range. Amino acid alterations of a human Fc region that result in such a desired binding activity can be found by comparing the human FcRn-binding activity in the acidic pH range and/or neutral pH range before and after amino acid alteration. Those skilled in the art can appropriately alter amino acids using known methods.


In the present invention, “alteration of amino acids” or “amino acid alteration” of an Fc region includes alteration into an amino acid sequence which is different from that of the starting Fc region. The starting Fc region may be any Fc region, as long as a variant modified from the starting Fc region can bind to human FcRn in an acidic pH range (i.e., the starting Fc region does not necessarily need to have an activity to bind to human FcRn in a neutral pH range). Examples of starting Fc regions preferably include Fc regions of IgG antibodies, i.e., native Fc regions. Furthermore, an altered Fc region modified from a starting Fc region which has been already modified can also be used preferably as an altered Fc region of the present invention. The “starting Fc region” can refer to the polypeptide itself, a composition comprising the starting Fc region, or an amino acid sequence encoding the starting Fc region. Starting Fc regions can comprise a known IgG antibody Fc region produced via recombination described briefly in section “Antibody”. The origin of starting Fc regions is not limited, and they may be obtained from human or any nonhuman organisms. Such organisms preferably include mice, rats, guinea pigs, hamsters, gerbils, cats, rabbits, dogs, goats, sheep, bovines, horses, camels and organisms selected from nonhuman primates. In another embodiment, starting Fc regions can also be obtained from cynomolgus monkeys, marmosets, rhesus monkeys, chimpanzees, or humans. Starting Fc regions can be obtained preferably from human IgG1; however, they are not limited to any particular IgG subclass. This means that an Fc region represented by human IgG1 (SEQ ID NO: 9), IgG2 (SEQ ID NO: 10), IgG3 (SEQ ID NO: 11), or IgG4 (SEQ ID NO: 12) can be used appropriately as a starting Fc region, and herein also means that an Fc region of an arbitrary IgG class or subclass derived from any organisms described above can be preferably used as a starting Fc region. Examples of naturally-occurring IgG variants or altered forms are described in published documents (Curr. Opin. Biotechnol. (2009) 20(6): 685-91; Curr. Opin. Immunol. (2008) 20(4), 460-470; Protein Eng. Des. Sel. (2010) 23(4): 195-202; International Publication Nos. WO 2009/086320, WO 2008/092117, WO 2007/041635, and WO 2006/105338); however, they are not limited to the examples.


Examples of alterations include those with one or more mutations, for example, mutations by substitution of different amino acid residues for amino acids of starting Fc regions, by insertion of one or more amino acid residues into starting Fc regions, or by deletion of one or more amino acids from starting Fc region. Preferably, the amino acid sequences of altered Fc regions comprise at least a part of the amino acid sequence of a non-native Fc region. Such variants necessarily have sequence identity or similarity less than 100% to their starting Fc region. In a preferred embodiment, the variants have amino acid sequence identity or similarity about 75% to less than 100%, more preferably about 80% to less than 100%, even more preferably about 85% to less than 100%, still more preferably about 90% to less than 100%, and yet more preferably about 95% to less than 100% to the amino acid sequence of their starting Fc region. In a non-limiting embodiment of the present invention, at least one amino acid is different between a modified Fc region of the present invention and its starting Fc region. Amino acid difference between a modified Fc region of the present invention and its starting Fc region can also be preferably specified based on amino acid differences at above-described particular amino acid positions according to EU numbering system.


Known methods such as site-directed mutagenesis (Kunkel et al. (Proc. Natl. Acad. Sci. USA (1985) 82, 488-492)) and Overlap extension PCR can be appropriately employed to alter the amino acids of Fc regions. Furthermore, various known methods can also be used as an amino acid alteration method for substituting amino acids by those other than natural amino acids (Annu Rev. Biophys. Biomol. Struct. (2006) 35, 225-249; Proc. Natl. Acad. Sci. U.S.A. (2003) 100 (11), 6353-6357). For example, a cell-free translation system (Clover Direct (Protein Express)) containing tRNAs in which amber suppressor tRNA, which is complementary to UAG codon (amber codon) that is a stop codon, is linked with an unnatural amino acid may be suitably used.


Fc regions comprised in the antigen-binding molecules of the present invention that have a human FcRn-binding activity in the acidic pH range can be obtained by any method. Specifically, one can screen for antigen-binding molecules comprising an Fc region of which human FcRn-binding activity in the acidic pH range is 20 μM (1(D) or stronger; in a more favorable embodiment, an Fc region of which human FcRn-binding activity in the acidic pH range is 2.0 μM (KD) or stronger; and in an even more favorable embodiment, an Fc region of which human FcRn-binding activity in the acidic pH range is 0.5 μM (KD) or stronger as a result of altering amino acids of an IgG-type human immunoglobulin used as a starting Fc region. Preferred Fc regions of IgG-type immunoglobulins for modification include, for example, those of human IgGs such as IgG1, IgG2, IgG3, and IgG4 shown in SEQ ID NOs: 9, 10, 11, and 12, respectively, and variants thereof.


When an antigen-binding molecule comprises the Fc region of human IgG1 as the Fc region, suitable examples of amino acids that may be altered to achieve the above-mentioned desired effects on FcRn binding under an acidic pH range condition by altering amino acids of an IgG-type human immunoglobulin as a starting Fc region, include at least one or more amino acids selected from the group of positions 238, 252, 253, 254, 255, 256, 265, 272, 286, 288, 303, 305, 307, 309, 311, 312, 317, 340, 356, 360, 362, 376, 378, 380, 382, 386, 388, 400, 413, 415, 424, 433, 434, 435, 436, 439, and 447 according to EU numbering as described in WO 2000/042072. Similarly, favorable examples of amino acids that allow such alteration include at least one or more amino acids selected from the group of positions 251, 252, 254, 255, 256, 308, 309, 311, 312, 385, 386, 387, 389, 428, 433, 434, and 436 according to EU numbering as described in WO 2002/060919. Furthermore, amino acids that allow such alteration include, for example, amino acids of positions 250, 314, and 428 according to EU numbering as described in WO2004/092219. Furthermore, favorable examples of amino acids that allow such alteration include at least one or more amino acids selected from the group of positions 251, 252, 307, 308, 378, 428, 430, 434, and 436 according to EU numbering as described in WO 2010/045193. Alteration of these amino acids enhances FcRn binding of the Fc region of an IgG-type immunoglobulin under an acidic pH range condition.


Fc regions having human FcRn-binding activity in the neutral pH range can also be obtained by altering amino acids of human immunoglobulin of IgG type used as the starting Fc region. The Fc regions of IgG type immunoglobulins adequate for modification include, for example, those of human IgGs such as IgG1, IgG2, IgG3, and IgG4 respectively represented by SEQ ID NOs: 9, 10, 11, and 12, and modified forms thereof. Amino acids of any positions may be altered into other amino acids, as long as the Fc regions have the human FcRn-binding activity in the neutral pH range or can increase the human FcRn-binding activity in the neutral range. When the antigen-binding molecule contains the Fc region of human IgG1 as the human Fc region, it is preferable that the resulting Fc region contains a modification that results in the effect of enhancing the human FcRn binding in the neutral pH range as compared to the binding activity of the starting Fc region of human IgG1. Amino acids that allow such alteration include, for example, one or more amino acids selected from the group of positions 221 to 225, 227, 228, 230, 232, 233 to 241, 243 to 252, 254 to 260, 262 to 272, 274, 276, 278 to 289, 291 to 312, 315 to 320, 324, 325, 327 to 339, 341, 343, 345, 360, 362, 370, 375 to 378, 380, 382, 385 to 387, 389, 396, 414, 416, 423, 424, 426 to 438, 440, and 442 according to EU numbering. Alteration of these amino acids enhances the human FcRn binding of the Fc region of IgG-type immunoglobulin in the neutral pH range.


From those described above, alterations that enhance the human FcRn binding in the neutral pH range are appropriately selected for use in the present invention. Particularly preferred amino acids of the modified Fc regions include, for example, amino acids at positions 237, 248, 250, 252, 254, 255, 256, 257, 258, 265, 286, 289, 297, 298, 303, 305, 307, 308, 309, 311, 312, 314, 315, 317, 332, 334, 360, 376, 380, 382, 384, 385, 386, 387, 389, 424, 428, 433, 434, and 436 according to the EU numbering system. The human FcRn-binding activity in the neutral pH range of the Fc region contained in an antigen-binding molecule can be increased by substituting at least one amino acid selected from the above amino acids into a different amino acid.


Particularly preferred alterations include, for example, at least one or more amino acids selected from the group of:


Met for the amino acid at position 237;


Ile for the amino acid at position 248;


Ala, Phe, Ile, Met, Gln, Ser, Val, Trp, or Tyr for the amino acid at position 250;


Phe, Trp, or Tyr for the amino acid at position 252;


Thr for the amino acid at position 254;


Glu for the amino acid at position 255;


Asp, Asn, Glu, or Gln for the amino acid at position 256;


Ala, Gly, Ile, Leu, Met, Asn, Ser, Thr, or Val for the amino acid at position 257;


His for the amino acid at position 258:


Ala for the amino acid at position 265;


Ala or Glu for the amino acid at position 286;


His for the amino acid at position 289;


Ala for the amino acid at position 297;


Ala for the amino acid at position 303;


Ala for the amino acid at position 305;


Ala, Asp, Phe, Gly, His, Ile, Lys, Leu, Met, Asn, Pro, Gln, Arg, Ser, Val, Trp, or Tyr for the amino acid at position 307;


Ala, Phe, Ile, Leu, Met, Pro, Gln, or Thr for the amino acid at position 308;


Ala, Asp, Glu, Pro, or Arg for the amino acid at position 309;


Ala, His, or Ile for the amino acid at position 311;


Ala or His for the amino acid at position 312;


Lys or Arg for the amino acid at position 314;


Ala, Asp, or His for the amino acid at position 315;


Ala for the amino acid at position 317;


Val for the amino acid at position 332;


Leu for the amino acid at position 334;


His for the amino acid at position 360;


Ala for the amino acid at position 376;


Ala for the amino acid at position 380;


Ala for the amino acid at position 382;


Ala for the amino acid at position 384;


Asp or His for the amino acid at position 385;


Pro for the amino acid at position 386;


Glu for the amino acid at position 387;


Ala or Ser for the amino acid at position 389;


Ala for the amino acid at position 424;


Ala, Asp, Phe, Gly, His, Ile, Lys, Leu, Asn, Pro, Gln, Ser, Thr, Val, Trp, or Tyr for the amino acid at position 428;


Lys for the amino acid at position 433;


Ala, Phe, His, Ser, Trp, or Tyr for the amino acid at position 434; and


His, Ile, Leu, Phe, Thr, or Val for the amino acid at position 436 of the Fc region according to EU numbering. Meanwhile, the number of amino acids to be altered is not particularly limited and amino acid at only one site may be altered and amino acids at two or more sites may be altered. Combinations of these amino acid alterations include, for example, the amino acid alterations shown in Tables 5-1 to 5-32.













TABLE 5-1







Variant
KD (M)
Site of amino acid alteration









F1
8.10E−07
N434W



F2
3.20E−06
M252Y/S254T/T256E



F3
2.50E−06
N434Y



F4
5.80E−06
N434S



F5
6.80E−06
N434A



F7
5.60E−06
M252Y



F8
4.20E−06
M252W



F9
1.40E−07
M252Y/S254T/T256E/N434Y



F10
6.90E−08
M252Y/S254T/T256E/N434W



F11
3.10E−07
M252Y/N434Y



F12
1.70E−07
M252Y/N434W



F13
3.20E−07
M252W/N434Y



F14
1.80E−07
M252W/N434W



F19
4.60E−07
P257L/N434Y



F20
4.60E−07
V308F/N434Y



F21
3.00E−08
M252Y/V308P/N434Y



F22
2.00E−06
M428L/N434S



F25
9.20E−09
M252Y/S254T/T256E/V308P/N434W



F26
1.00E−06
I332V



F27
7.40E−06
G237M



F29
1.40E−06
I332V/N434Y



F31
2.80E−06
G237M/V308F



F32
8.00E−07
S254T/N434W



F33
2.30E−06
S254T/N434Y



F34
2.80E−07
T256E/N434W



F35
8.40E−07
T256E/N434Y



F36
3.60E−07
S254T/T256E/N434W



F37
1.10E−06
S254T/T256E/N434Y



F38
1.00E−07
M252Y/S254T/N434W



F39
3.00E−07
M252Y/S254T/N434Y



F40
8.20E−08
M252Y/T256E/N434W



F41
1.50E−07
M252Y/T256E/N434Y










Table 5-2 is a continuation table of Table 5-1.













TABLE 5-2









F42
1.00E−06
M252Y/S254T/T256E/N434A



F43
1.70E−06
M252Y/N434A



F44
1.10E−06
M252W/N434A



F47
2.40E−07
M252Y/T256Q/N434W



F48
3.20E−07
M252Y/T256Q/N434Y



F49
5.10E−07
M252F/T256D/N434W



F50
1.20E−06
M252F/T256D/N434Y



F51
8.10E−06
N434F/Y436H



F52
3.10E−06
H433K/N434F/Y436H



F53
1.00E−06
I332V/N434W



F54
8.40E−08
V308P/N434W



F56
9.40E−07
I332V/M428L/N434Y



F57
1.10E−05
G385D/Q386P/N389S



F58
7.70E−07
G385D/Q386P/N389S/N434W



F59
2.40E−06
G385D/Q386P/N389S/N434Y



F60
1.10E−05
G385H



F61
9.70E−07
G385H/N434W



F62
1.90E−06
G385H/N434Y



F63
2.50E−06
N434F



F64
5.30E−06
N434H



F65
2.90E−07
M252Y/S254T/T256E/N434F



F66
4.30E−07
M252Y/S254T/T256E/N434H



F67
6.30E−07
M252Y/N434F



F68
9.30E−07
M252Y/N434H



F69
5.10E−07
M428L/N434W



F70
1.50E−06
M428L/N434Y



F71
8.30E−08
M252Y/S254T/T256E/M428L/N434W



F72
2.00E−07
M252Y/S254T/T256E/M428L/N434Y



F73
1.70E−07
M252Y/M428L/N434W



F74
4.60E−07
M252Y/M428L/N434Y



F75
1.40E−06
M252Y/M428L/N434A



F76
1.00E−06
M252Y/S254T/T256E/M428L/N434A



F77
9.90E−07
T256E/M428L/N434Y



F78
7.80E−07
S254T/M428L/N434W










Table 5-3 is a continuation table of Table 5-2.











TABLE 5-3







F79
5.90E−06
S254T/T256E/N434A


F80
2.70E−06
M252Y/T256Q/N434A


F81
1.60E−06
M252Y/T256E/N434A


F82
1.10E−06
T256Q/N434W


F83
2.60E−06
T256Q/N434Y


F84
2.80E−07
M252W/T256Q/N434W


F85
5.50E−07
M252W/T256Q/N434Y


F86
1.50E−06
S254T/T256Q/N434W


F87
4.30E−06
S254T/T256Q/N434Y


F88
1.90E−07
M252Y/S254T/T256Q/N434W


F89
3.60E−07
M252Y/S254T/T256Q/N434Y


F90
1.90E−08
M252Y/T256E/V308P/N434W


F91
4.80E−08
M252Y/V308P/M428L/N434Y


F92
1.10E−08
M252Y/S254T/T256E/V308P/M428L/N434W


F93
7.40E−07
M252W/M428L/N434W


F94
3.70E−07
P257L/M428L/N434Y


F95
2.60E−07
M252Y/S254T/T256E/M428L/N434F


F99
6.20E−07
M252Y/T256E/N434H


F101
1.10E−07
M252W/T256Q/P257L/N434Y


F103
4.40E−08
P238A/M252Y/V308P/N434Y


F104
3.70E−08
M252Y/D265A/V308P/N434Y


F105
7.50E−08
M252Y/T307A/V308P/N434Y


F106
3.70E−08
M252Y/V303A/V308P/N434Y


F107
3.40E−08
M252Y/V308P/D376A/N434Y


F108
4.10E−08
M252Y/V305A/V308P/N434Y


F109
3.20E−08
M252Y/V308P/Q311A/N434Y


F111
3.20E−08
M252Y/V308P/K317A/N434Y


F112
6.40E−08
M252Y/V308P/E380A/N434Y


F113
3.20E−08
M252Y/V308P/E382A/N434Y


F114
3.80E−08
M252Y/V308P/S424A/N434Y


F115
6.60E−06
T307A/N434A


F116
8.70E−06
E380A/N434A


F118
1.40E−05
M428L


F119
5.40E−06
T250Q/M428L









Table 5-4 is a continuation table of Table 5-3.











TABLE 5-4







F120
6.30E−08
P257L/V308P/M428L/N434Y


F121
1.50E−08
M252Y/T256E/V308P/M428L/N434W


F122
1.20E−07
M252Y/T256E/M428L/N434W


F123
3.00E−08
M252Y/T256E/V308P/N434Y


F124
2.90E−07
M252Y/T256E/M428L/N434Y


F125
2.40E−08
M252Y/S254T/T256E/V308P/M428L/N434Y


F128
1.70E−07
P257L/M428L/N434W


F129
2.20E−07
P257A/M428L/N434Y


F131
3.00E−06
P257G/M428L/N434Y


F132
2.10E−07
P257I/M428L/N434Y


F133
4.10E−07
P257M/M428L/N434Y


F134
2.70E−07
P257N/M428L/N434Y


F135
7.50E−07
P257S/M428L/N434Y


F136
3.80E−07
P257T/M428L/N434Y


F137
4.60E−07
P257V/M428L/N434Y


F139
1.50E−08
M252W/V308P/N434W


F140
3.60E−08
S239K/M252Y/V308P/N434Y


F141
3.50E−08
M252Y/S298G/V308P/N434Y


F142
3.70E−08
M252Y/D270F/V308P/N434Y


F143
2.00E−07
M252Y/V308A/N434Y


F145
5.30E−08
M252Y/V308F/N434Y


F147
2.40E−07
M252Y/V308I/N434Y


F149
1.90E−07
M252Y/V308L/N434Y


F150
2.00E−07
M252Y/V308M/N434Y


F152
2.70E−07
M252Y/V308Q/N434Y


F154
1.80E−07
M252Y/V308T/N434Y


F157
1.50E−07
P257A/V308P/M428L/N434Y


F158
5.90E−08
P257T/V308P/M428L/N434Y


F159
4.40E−08
P257V/V308P/M428L/N434Y


F160
8.50E−07
M252W/M428I/N434Y


F162
1.60E−07
M252W/M428Y/N434Y


F163
4.20E−07
M252W/M428F/N434Y


F164
3.70E−07
P238A/M252W/N434Y


F165
2.90E−07
M252W/D265A/N434Y









Table 5-5 is a continuation table of Table 5-4.













TABLE 5-5









F166
1.50E−07
M252W/T307Q/N434Y



F167
2.90E−07
M252W/V303A/N434Y



F168
3.20E−07
M252W/D376A/N434Y



F169
2.90E−07
M252W/V305A/N434Y



F170
1.70E−07
M252W/Q311A/N434Y



F171
1.90E−07
M252W/D312A/N434Y



F172
2.20E−07
M252W/K317A/N434Y



F173
7.70E−07
M252W/E380A/N434Y



F174
3.40E−07
M252W/E382A/N434Y



F175
2.70E−07
M252W/S424A/N434Y



F176
2.90E−07
S239K/M252W/N434Y



F177
2.80E−07
M252W/S298G/N434Y



F178
2.70E−07
M252W/D270F/N434Y



F179
3.10E−07
M252W/N325G/N434Y



F182
6.60E−08
P257A/M428L/N434W



F183
2.20E−07
P257T/M428L/N434W



F184
2.70E−07
P257V/M428L/N434W



F185
2.60E−07
M252W/I332V/N434Y



F188
3.00E−06
P257I/Q311I



F189
1.90E−07
M252Y/T307A/N434Y



F190
1.10E−07
M252Y/T307Q/N434Y



F191
1.60E−07
P257L/T307A/M428L/N434Y



F192
1.10E−07
P257A/T307A/M428L/N434Y



F193
8.50E−08
P257T/T307A/M428L/N434Y



F194
1.20E−07
P257V/T307A/M428L/N434Y



F195
5.60E−08
P257L/T307Q/M428L/N434Y



F196
3.50E−08
P257A/T307Q/M428L/N434Y



F197
3.30E−08
P257T/T307Q/M428L/N434Y



F198
4.80E−08
P257V/T307Q/M428L/N434Y



F201
2.10E−07
M252Y/T307D/N434Y



F203
2.40E−07
M252Y/T307F/N434Y



F204
2.10E−07
M252Y/T307G/N434Y



F205
2.00E−07
M252Y/T307H/N434Y



F206
2.30E−07
M252Y/T307I/N434Y










Table 5-6 is a continuation table of Table 5-5.













TABLE 5-6









F207
9.40E−07
M252Y/T307K/N434Y



F208
3.90E−07
M252Y/T307L/N434Y



F209
1.30E−07
M252Y/T307M/N434Y



F210
2.90E−07
M252Y/T307N/N434Y



F211
2.40E−07
M252Y/T307P/N434Y



F212
6.80E−07
M252Y/T307R/N434Y



F213
2.30E−07
M252Y/T307S/N434Y



F214
1.70E−07
M252Y/T307V/N434Y



F215
9.60E−08
M252Y/T307W/N434Y



F216
2.30E−07
M252Y/T307Y/N434Y



F217
2.30E−07
M252Y/K334L/N434Y



F218
2.60E−07
M252Y/G385H/N434Y



F219
2.50E−07
M252Y/T289H/N434Y



F220
2.50E−07
M252Y/Q311H/N434Y



F221
3.10E−07
M252Y/D312H/N434Y



F222
3.40E−07
M252Y/N315H/N434Y



F223
2.70E−07
M252Y/K360H/N434Y



F225
1.50E−06
M252Y/L314R/N434Y



F226
5.40E−07
M252Y/L314K/N434Y



F227
1.20E−07
M252Y/N286E/N434Y



F228
2.30E−07
M252Y/L309E/N434Y



F229
5.10E−07
M252Y/R255E/N434Y



F230
2.50E−07
M252Y/P387E/N434Y



F236
8.90E−07
K248I/M428L/N434Y



F237
2.30E−07
M252Y/M428A/N434Y



F238
7.40E−07
M252Y/M428D/N434Y



F240
7.20E−07
M252Y/M428F/N434Y



F241
1.50E−06
M252Y/M428G/N434Y



F242
8.50E−07
M252Y/M428H/N434Y



F243
1.80E−07
M252Y/M428I/N434Y



F244
1.30E−06
M252Y/M428K/N434Y



F245
4.70E−07
M252Y/M428N/N434Y



F246
1.10E−06
M252Y/M428P/N434Y



F247
4.40E−07
M252Y/M428Q/N434Y










Table 5-7 is a continuation table of Table 5-6.













TABLE 5-7









F249
6.40E−07
M252Y/M428S/N434Y



F250
2.90E−07
M252Y/M428T/N434Y



F251
1.90E−07
M252Y/M428V/N434Y



F252
1.00E−06
M252Y/M428W/N434Y



F253
7.10E−07
M252Y/M428Y/N434Y



F254
7.50E−08
M252W/T307Q/M428Y/N434Y



F255
1.10E−07
M252W/Q311A/M428Y/N434Y



F256
5.40E−08
M252W/T307Q/Q311A/M428Y/N434Y



F257
5.00E−07
M252Y/T307A/M428Y/N434Y



F258
3.20E−07
M252Y/T307Q/M428Y/N434Y



F259
2.80E−07
M252Y/D270F/N434Y



F260
1.30E−07
M252Y/T307A/Q311A/N434Y



F261
8.40E−08
M252Y/T307Q/Q311A/N434Y



F262
1.90E−07
M252Y/T307A/Q311H/N434Y



F263
1.10E−07
M252Y/T307Q/Q311H/N434Y



F264
2.80E−07
M252Y/E382A/N434Y



F265
6.80E−07
M252Y/E382A/M428Y/N434Y



F266
4.70E−07
M252Y/T307A/E382A/M428Y/N434Y



F267
3.20E−07
M252Y/T307Q/E382A/M428Y/N434Y



F268
6.30E−07
P238A/M252Y/M428F/N434Y



F269
5.20E−07
M252Y/V305A/M428F/N434Y



F270
6.60E−07
M252Y/N325G/M428F/N434Y



F271
6.90E−07
M252Y/D376A/M428F/N434Y



F272
6.80E−07
M252Y/E380A/M428F/N434Y



F273
6.50E−07
M252Y/E382A/M428F/N434Y



F274
7.60E−07
M252Y/E380A/E382A/M428F/N434Y



F275
4.20E−08
S239K/M252Y/V308P/E382A/N434Y



F276
4.10E−08
M252Y/D270F/V308P/E382A/N434Y



F277
1.30E−07
S239K/M252Y/V308P/M428Y/N434Y



F278
3.00E−08
M252Y/T307Q/V308P/E382A/N434Y



F279
6.10E−08
M252Y/V308P/Q311H/E382A/N434Y



F280
4.10E−08
S239K/M252Y/D270F/V308P/N434Y



F281
9.20E−08
M252Y/V308P/E382A/M428F/N434Y



F282
2.90E−08
M252Y/V308P/E382A/M428L/N434Y










Table 5-8 is a continuation table of Table 5-7.











TABLE 5-8







F283
1.00E−07
M252Y/V308P/E382A/M428Y/N434Y


F284
1.00E−07
M252Y/V308P/M428Y/N434Y


F285
9.90E−08
M252Y/V308P/M428F/N434Y


F286
1.20E−07
S239K/M252Y/V308P/E382A/M428Y/N434Y


F287
1.00E−07
M252Y/V308P/E380A/E382A/M428F/N434Y


F288
1.90E−07
M252Y/T256E/E382A/N434Y


F289
4.80E−07
M252Y/T256E/M428Y/N434Y


F290
4.60E−07
M252Y/T256E/E382A/M428Y/N434Y


F292
2.30E−08
S239K/M252Y/V308P/E382A/M428I/N434Y


F293
5.30E−08
M252Y/V308P/E380A/E382A/M428I/N434Y


F294
1.10E−07
S239K/M252Y/V308P/M428F/N434Y


F295
6.80E−07
S239K/M252Y/E380A/E382A/M428F/N434Y


F296
4.90E−07
M252Y/Q311A/M428Y/N434Y


F297
5.10E−07
M252Y/D312A/M428Y/N434Y


F298
4.80E−07
M252Y/Q311A/D312A/M428Y/N434Y


F299
9.40E−08
S239K/M252Y/V308P/Q311A/M428Y/N434Y


F300
8.30E−08
S239K/M252Y/V308P/D312A/M428Y/N434Y


F301
7.20E−08
S239K/M252Y/V308P/Q311A/D312A/M428Y/N434Y


F302
1.90E−07
M252Y/T256E/T307P/N434Y


F303
6.70E−07
M252Y/T307P/M428Y/N434Y


F304
1.60E−08
M252W/V308P/M428Y/N434Y


F305
2.70E−08
M252Y/T256E/V308P/E382A/N434Y


F306
3.60E−08
M252W/V308P/E382A/N434Y


F307
3.60E−08
S239K/M252W/V308P/E382A/N434Y


F308
1.90E−08
S239K/M252W/V308P/E382A/M428Y/N434Y


F310
9.40E−08
S239K/M252W/V308P/E382A/M428I/N434Y


F311
2.80E−08
S239K/M252W/V308P/M428F/N434Y


F312
4.50E−07
S239K/M252W/E380A/E382A/M428F/N434Y


F313
6.50E−07
S239K/M252Y/T307P/M428Y/N434Y


F314
3.20E−07
M252Y/T256E/Q311A/D312A/M428Y/N434Y


F315
6.80E−07
S239K/M252Y/M428Y/N434Y


F316
7.00E−07
S239K/M252Y/D270F/M428Y/N434Y


F317
1.10E−07
S239K/M252Y/D270F/V308P/M428Y/N434Y


F318
1.80E−08
S239K/M252Y/V308P/M428I/N434Y









Table 5-9 is a continuation table of Table 5-8.











TABLE 5-9







F320
2.00E−08
S239K/M252Y/V308P/N325G/E382A/M428I/N434Y


F321
3.20E−08
S239K/M252Y/D270F/V308P/N325G/N434Y


F322
9.20E−08
S239K/M252Y/D270F/T307P/V308P/N434Y


F323
2.70E−08
S239K/M252Y/T256E/D270F/V308P/N434Y


F324
2.80E−08
S239K/M252Y/D270F/T307Q/V308P/N434Y


F325
2.10E−08
S239K/M252Y/D270F/T307Q/V308P/Q311A/N434Y


F326
7.50E−08
S239K/M252Y/D270F/T307Q/Q311A/N434Y


F327
6.50E−08
S239K/M252Y/T256E/D270F/T307Q/Q311A/N434Y


F328
1.90E−08
S239K/M252Y/D270F/V308P/M428I/N434Y


F329
1.20E−08
S239K/M252Y/D270F/N286E/V308P/N434Y


F330
3.60E−08
S239K/M252Y/D270F/V308P/L309E/N434Y


F331
3.00E−08
S239K/M252Y/D270F/V308P/P387E/N434Y


F333
7.40E−08
S239K/M252Y/D270F/T307Q/L309E/Q311A/N434Y


F334
1.90E−08
S239K/M252Y/D270F/V308P/N325G/M428I/N434Y


F335
1.50E−08
S239K/M252Y/T256E/D270F/V308P/M428I/N434Y


F336
1.40E−08
S239K/M252Y/D270F/T307Q/V308P/Q311A/M428I/




N434Y


F337
5.60E−08
S239K/M252Y/D270F/T307Q/Q311A/M428I/N434Y


F338
7.70E−09
S239K/M252Y/D270F/N286E/V308P/M428I/N434Y


F339
1.90E−08
S239K/M252Y/D270E/V308P/L309E/M428I/N434Y


F343
3.20E−08
S239K/M252Y/D270F/V308P/M428L/N434Y


F344
3.00E−08
S239K/M252Y/V308P/M428L/N434Y


F349
1.50E−07
S239K/M252Y/V308P/L309P/M428L/N434Y


F350
1.70E−07
S239K/M252Y/V308P/L309R/M428L/N434Y


F352
6.00E−07
S239K/M252Y/L309P/M428L/N434Y


F353
1.10E−06
S239K/M252Y/L309R/M428L/N434Y


F354
2.80E−08
S239K/M252Y/T307Q/V308P/M428L/N434Y


F356
3.40E−08
S239K/M252Y/D270F/V308P/L309E/P387E/N434Y


F357
1.60E−08
S239K/M252Y/T256E/D270F/V308P/N325G/M428I/




N434Y


F358
1.00E−07
S239K/M252Y/T307Q/N434Y


F359
4.20E−07
P257V/T307Q/M428I/N434Y


F360
1.30E−06
P257V/T307Q/M428V/N434Y


F362
5.40E−08
P257V/T307Q/N325G/M428L/N434Y


F363
4.10E−08
P257V/T307Q/Q311A/M428L/N434Y


F364
3.50E−08
P257V/T307Q/Q311A/N325G/M428L/N434Y









Table 5-10 is a continuation table of Table 5-9.











TABLE 5-10







F365
5.10E−08
P257V/V305A/T307Q/M428L/N434Y


F367
1.50E−08
S239K/M252Y/E258H/D270F/T307Q/V308P/Q311A/N434Y


F368
2.00E−08
S239K/M252Y/D270F/V308P/N325G/E382A/M428I/N434Y


F369
7.50E−08
M252Y/P257V/T307Q/M428I/N434Y


F372
1.30E−08
S239K/M252W/V308P/M428Y/N434Y


F373
1.10E−08
S239K/M252W/V308P/Q311A/M428Y/N434Y


F374
1.20E−08
S239K/M252W/T256E/V308P/M428Y/N434Y


F375
5.50E−09
S239K/M252W/N286E/V308P/M428Y/N434Y


F376
9.60E−09
S239K/M252Y/T256E/D270F/N286E/V308P/N434Y


F377
1.30E−07
S239K/M252W/T307P/M428Y/N434Y


F379
9.00E−09
S239K/M252W/T256E/V308P/Q311A/M428Y/N434Y


F380
5.60E−09
S239K/M252W/T256E/N286E/V308P/M428Y/N434Y


F381
1.10E−07
P257V/T307A/Q311A/M428L/N434Y


F382
8.70E−08
P257V/V305A/T307A/M428L/N434Y


F386
3.20E−08
M252Y/V308P/L309E/N434Y


F387
1.50E−07
M252Y/V308P/L309D/N434Y


F388
7.00E−08
M252Y/V308P/L309A/N434Y


F389
1.70E−08
M252W/V308P/L309E/M428Y/N434Y


F390
6.80E−08
M252W/V308P/L309D/M428Y/N434Y


F391
3.60E−08
M252W/V308P/L309A/M428Y/N434Y


F392
6.90E−09
S239K/M252Y/N286E/V308P/M428I/N434Y


F393
1.20E−08
S239K/M252Y/N286E/V308P/N434Y


F394
5.30E−08
S239K/M252Y/T307Q/Q311A/M428I/N434Y


F395
2.40E−08
S239K/M252Y/T256E/V308P/N434Y


F396
2.00E−08
S239K/M252Y/D270F/N286E/T307Q/Q311A/M428I/N434Y


F397
4.50E−08
S239K/M252Y/D270F/T307Q/Q311A/P387E/M428I/N434Y


F398
4.40E−09
S239K/M252Y/D270F/N286E/T307Q/V308P/Q311A/M428I/N434Y


F399
6.50E−09
S239K/M252Y/D270F/N286E/T307Q/V308P/M428I/N434Y


F400
6.10E−09
S239K/M252Y/D270F/N286E/V308P/Q311A/M428I/N434Y


F401
6.90E−09
S239K/M252Y/D270F/N286E/V308P/P387E/M428I/N434Y


F402
2.30E−08
P257V/T307Q/M428L/N434W


F403
5.10E−08
P257V/T307A/M428L/N434W


F404
9.40E−08
P257A/T307Q/L309P/M428L/N434Y


F405
1.70E−07
P257V/T307Q/L309P/M428L/N434Y









Table 5-11 is a continuation table of Table 5-10.











TABLE 5-11







F406
1.50E−07
P257A/T307Q/L309R/M428L/N434Y


F407
1.60E−07
P257V/T307Q/L309R/M428L/N434Y


F408
2.50E−07
P257V/N286E/M428L/N434Y


F409
2.00E−07
P257V/P387E/M428L/N434Y


F410
2.20E−07
P257V/T307H/M428L/N434Y


F411
1.30E−07
P257V/T307N/M428L/N434Y


F412
8.80E−08
P257V/T307G/M428L/N434Y


F413
1.20E−07
P257V/T307P/M428L/N434Y


F414
1.10E−07
P257V/T307S/M428L/N434Y


F415
5.60E−08
P257V/N286E/T307A/M428L/N434Y


F416
9.40E−08
P257V/T307A/P387E/M428L/N434Y


F418
6.20E−07
S239K/M252Y/T307P/N325G/M428Y/N434Y


F419
1.60E−07
M252Y/T307A/Q311H/K360H/N434Y


F420
1.50E−07
M252Y/T307A/Q311H/P387E/N434Y


F421
1.30E−07
M252Y/T307A/Q311H/M428A/N434Y


F422
1.80E−07
M252Y/T307A/Q311H/E382A/N434Y


F423
8.40E−08
M252Y/T307W/Q311H/N434Y


F424
9.40E−08
S239K/P257A/V308P/M428L/N434Y


F425
8.00E−08
P257A/V308P/L309E/M428L/N434Y


F426
8.40E−08
P257V/T307Q/N434Y


F427
1.10E−07
M252Y/P257V/T307Q/M428V/N434Y


F428
8.00E−08
M252Y/P257V/T307Q/M428L/N434Y


F429
3.70E−08
M252Y/P257V/T307Q/N434Y


F430
8.10E−08
M252Y/P257V/T307Q/M428Y/N434Y


F431
6.50E−08
M252Y/P257V/T307Q/M428F/N434Y


F432
9.20E−07
P257V/T307Q/Q311A/N325G/M428V/N434Y


F433
6.00E−08
P257V/T307Q/Q311A/N325G/N434Y


F434
2.00E−08
P257V/T307Q/Q311A/N325G/M428Y/N434Y


F435
2.50E−08
P257V/T307Q/Q311A/N325G/M428F/N434Y


F436
2.50E−07
P257A/T307Q/M428V/N434Y


F437
5.70E−08
P257A/T307Q/N434Y


F438
3.60E−08
P257A/T307Q/M428Y/N434Y


F439
4.00E−08
P257A/T307Q/M428F/N434Y


F440
1.50E−08
P257V/N286E/T307Q/Q311A/N325G/M428L/N434Y









Table 5-12 is a continuation table of Table 5-11.











TABLE 5-12







F441
1.80E−07
P257A/Q311A/M428L/N434Y


F442
2.00E−07
P257A/Q311H/M428L/N434Y


F443
5.50E−08
P257A/T307Q/Q311A/M428L/N434Y


F444
1.40E−07
P257A/T307A/Q311A/M428L/N434Y


F445
6.20E−08
P257A/T307Q/Q311H/M428L/N434Y


F446
1.10E−07
P257A/T307A/Q311H/M428L/N434Y


F447
1.40E−08
P257A/N286E/T307Q/M428L/N434Y


F448
5.30E−08
P257A/N286E/T307A/M428L/N434Y


F449
5.70E−07
S239K/M252Y/D270F/T307P/N325G/M428Y/N434Y


F450
5.20E−07
S239K/M252Y/T307P/L309E/N325G/M428Y/N434Y


F451
1.00E−07
P257S/T307A/M428L/N434Y


F452
1.40E−07
P257M/T307A/M428L/N434Y


F453
7.80E−08
P257N/T307A/M428L/N434Y


F454
9.60E−08
P257I/T307A/M428L/N434Y


F455
2.70E−08
P257V/T307Q/M428Y/N434Y


F456
3.40E−08
P257V/T307Q/M428F/N434Y


F457
4.00E−08
S239K/P257V/V308P/M428L/N434Y


F458
1.50E−08
P257V/T307Q/V308P/N325G/M428L/N434Y


F459
1.30E−08
P257V/T307Q/V308P/Q311A/N325G/M428L/N434Y


F460
4.70E−08
P257V/T307A/V308P/N325G/M428L/N434Y


F462
8.50E−08
P257A/V308P/N325G/M428L/N434Y


F463
1.30E−07
P257A/T307A/V308P/M428L/N434Y


F464
5.50E−08
P257A/T307Q/V308P/M428L/N434Y


F465
2.10E−08
P257V/N286E/T307Q/N325G/M428L/N434Y


F466
3.50E−07
T256E/P257V/N434Y


F467
5.70E−07
T256E/P257T/N434Y


F468
5.70E−08
S239K/P257T/V308P/M428L/N434Y


F469
5.60E−08
P257T/V308P/N325G/M428L/N434Y


F470
5.40E−08
T256E/P257T/V308P/N325G/M428L/N434Y


F471
6.60E−08
P257T/V308P/N325G/E382A/M428L/N434Y


F472
5.40E−08
P257T/V308P/N325G/P387E/M428L/N434Y


F473
4.50E−07
P257T/V308P/L309P/N325G/M428L/N434Y


F474
3.50E−07
P257T/V308P/L309R/N325G/M428L/N434Y


F475
4.30E−08
T256E/P257V/T307Q/M428L/N434Y









Table 5-13 is a continuation table of Table 5-12.











TABLE 5-13







F476
5.50E−08
P257V/T307Q/E382A/M428L/N434Y


F477
4.30E−08
P257V/T307Q/P387E/M428L/N434Y


F480
3.90E−08
P257L/V308P/N434Y


F481
5.60E−08
P257T/T307Q/N434Y


F482
7.00E−08
P257V/T307Q/N325G/N434Y


F483
5.70E−08
P257V/T307Q/Q311A/N434Y


F484
6.20E−08
P257V/V305A/T307Q/N434Y


F485
9.70E−08
P257V/N286E/T307A/N434Y


F486
3.40E−07
P257V/T307Q/L309R/Q311H/M428L/N434Y


F488
3.50E−08
P257V/V308P/N325G/M428L/N434Y


F490
7.50E−08
S239K/P257V/V308P/Q311H/M428L/N434Y


F492
9.80E−08
P257V/V305A/T307A/N325G/M428L/N434Y


F493
4.90E−07
S239K/D270F/T307P/N325G/M428Y/N434Y


F497
3.10E−06
P257T/T307A/M428V/N434Y


F498
1.30E−06
P257A/M428V/N434Y


F499
5.20E−07
P257A/T307A/M428V/N434Y


F500
4.30E−08
P257S/T307Q/M428L/N434Y


F506
1.90E−07
P257V/N297A/T307Q/M428L/N434Y


F507
5.10E−08
P257V/N286A/T307Q/M428L/N434Y


F508
1.10E−07
P257V/T307Q/N315A/M428L/N434Y


F509
5.80E−08
P257V/T307Q/N384A/M428L/N434Y


F510
5.30E−08
P257V/T307Q/N389A/M428L/N434Y


F511
4.20E−07
P257V/N434Y


F512
5.80E−07
P257T/N434Y


F517
3.10E−07
P257V/N286E/N434Y


F518
4.20E−07
P257T/N286E/N434Y


F519
2.60E−08
P257V/N286E/T307Q/N434Y


F521
1.10E−08
P257V/N286E/T307Q/M428Y/N434Y


F523
2.60E−08
P257V/V305A/T307Q/M428Y/N434Y


F526
1.90E−08
P257T/T307Q/M428Y/N434Y


F527
9.40E−09
P257V/T307Q/V308P/N325G/M428Y/N434Y


F529
2.50E−08
P257T/T307Q/M428F/N434Y


F533
1.20E−08
P257A/N286E/T307Q/M428F/N434Y


F534
1.20E−08
P257A/N286E/T307Q/M428Y/N434Y









Table 5-14 is a continuation table of Table 5-13.













TABLE 5-14









F535
3.90E−08
T250A/P257V/T307Q/M428L/N434Y



F538
9.90E−08
T250F/P257V/T307Q/M428L/N434Y



F541
6.00E−08
T250I/P257V/T307Q/M428L/N434Y



F544
3.10E−08
T250M/P257V/T307Q/M428L/N434Y



F549
5.40E−08
T250S/P257V/T307Q/M428L/N434Y



F550
5.90E−08
T250V/P257V/T307Q/M428L/N434Y



F551
1.20E−07
T250W/P257V/T307Q/M428L/N434Y



F552
1.10E−07
T250Y/P257V/T307Q/M428L/N434Y



F553
1.70E−07
M252Y/Q311A/N434Y



F554
2.80E−08
S239K/M252Y/S254T/V308P/N434Y



F556
1.50E−06
M252Y/T307Q/Q311A



F559
8.00E−08
M252Y/S254T/N286E/N434Y



F560
2.80E−08
M252Y/S254T/V308P/N434Y



F561
1.40E−07
M252Y/S254T/T307A/N434Y



F562
8.30E−08
M252Y/S254T/T307Q/N434Y



F563
1.30E−07
M252Y/S254T/Q311A/N434Y



F564
1.90E−07
M252Y/S254T/Q311H/N434Y



F565
9.20E−08
M252Y/S254T/T307A/Q311A/N434Y



F566
6.10E−08
M252Y/S254T/T307Q/Q311A/N434Y



F567
2.20E−07
M252Y/S254T/M428I/N434Y



F568
1.10E−07
M252Y/T256E/T307A/Q311H/N434Y



F569
2.00E−07
M252Y/T256Q/T307A/Q311H/N434Y



F570
1.30E−07
M252Y/S254T/T307A/Q311H/N434Y



F571
8.10E−08
M252Y/N286E/T307A/Q311H/N434Y



F572
1.00E−07
M252Y/T307A/Q311H/M428I/N434Y



F576
1.60E−06
M252Y/T256E/T307Q/Q311H



F577
1.30E−06
M252Y/N286E/T307A/Q311A



F578
5.70E−07
M252Y/N286E/T307Q/Q311A



F580
8.60E−07
M252Y/N286E/T307Q/Q311H



F581
7.20E−08
M252Y/T256E/N286E/N434Y



F582
7.50E−07
S239K/M252Y/V308P



F583
7.80E−07
S239K/M252Y/V308P/E382A



F584
6.30E−07
S239K/M252Y/T256E/V308P



F585
2.90E−07
S239K/M252Y/N286E/V308P










Table 5-15 is a continuation table of Table 5-14.











TABLE 5-15







F586
1.40E−07
S239K/M252Y/N286E/V308P/M428I


F587
1.90E−07
M252Y/N286E/M428L/N434Y


F592
2.00E−07
M252Y/S254T/E382A/N434Y


F593
3.10E−08
S239K/M252Y/S254T/V308P/M428I/N434Y


F594
1.60E−08
S239K/M252Y/T256E/V308P/M428I/N434Y


F595
1.80E−07
S239K/M252Y/M428I/N434Y


F596
4.00E−07
M252Y/D312A/E382A/M428Y/N434Y


F597
2.20E−07
M252Y/E382A/P387E/N434Y


F598
1.40E−07
M252Y/D312A/P387E/N434Y


F599
5.20E−07
M252Y/P387E/M428Y/N434Y


F600
2.80E−07
M252Y/T256Q/E382A/N434Y


F601
9.60E−09
M252Y/N286E/V308P/N434Y


F608

G236A/S239D/I332E


F611
2.80E−07
M252Y/V305T/T307P/V308I/L309A/N434Y


F612
3.60E−07
M252Y/T307P/V308I/L309A/N434Y


F613

S239D/A330L/I332E


F616

S239D/K326D/L328Y


F617
7.40E−07
S239K/N434W


F618
6.40E−07
S239K/V308F/N434Y


F619
3.10E−07
S239K/M252Y/N434Y


F620
2.10E−07
S239K/M252Y/S254T/N434Y


F621
1.50E−07
S239K/M252Y/T307A/Q311H/N434Y


F622
3.50E−07
S239K/M252Y/T256Q/N434Y


F623
1.80E−07
S239K/M252W/N434W


F624
1.40E−08
S239K/P257A/N286E/T307Q/M428L/N434Y


F625
7.60E−08
S239K/P257A/T307Q/M428L/N434Y


F626
1.30E−06
V308P


F629
3.90E−08
M252Y/V279L/V308P/N434Y


F630
3.70E−08
S239K/M252Y/V279L/V308P/N434Y


F633
2.40E−08
M252Y/V282D/V308P/N434Y


F634
3.20E−08
S239K/M252Y/V282D/V308P/N434Y


F635
4.50E−08
M252Y/V284K/V308P/N434Y


F636
4.80E−08
S239K/M252Y/V284K/V308P/N434Y


F637
1.50E−07
M252Y/K288S/V308P/N434Y









Table 5-16 is a continuation table of Table 5-15.











TABLE 5-16







F638
1.40E−07
S239K/M252Y/K288S/V308P/N434Y


F639
2.70E−08
M252Y/V308P/G385R/N434Y


F640
3.60E−08
S239K/M252Y/V308P/G385R/N434Y


F641
3.00E−08
M252Y/V308P/Q386K/N434Y


F642
3.00E−08
S239K/M252Y/V308P/Q386K/N434Y


F643
3.20E−08
L235G/G236R/S239K/M252Y/V308P/N434Y


F644
3.00E−08
G236R/S239K/M252Y/V308P/N434Y


F645
3.30E−08
S239K/M252Y/V308P/L328R/N434Y


F646
3.80E−08
S239K/M252Y/N297A/V308P/N434Y


F647
2.90E−08
P238D/M252Y/V308P/N434Y


F648

P238D


F649
1.20E−07
S239K/M252Y/N286E/N434Y


F650
1.70E−07
S239K/M252Y/T256E/N434Y


F651
1.80E−07
S239K/M252Y/Q311A/N434Y


F652
2.40E−07
P238D/M252Y/N434Y


F654
3.20E−08
L235K/S239K/M252Y/V308P/N434Y


F655
3.40E−08
L235R/S239K/M252Y/V308P/N434Y


F656
3.30E−08
G237K/S239K/M252Y/V308P/N434Y


F657
3.20E−08
G237R/S239K/M252Y/V308P/N434Y


F658
3.20E−08
P238K/S239K/M252Y/V308P/N434Y


F659
3.00E−08
P238R/S239K/M252Y/V308P/N434Y


F660
3.10E−08
S239K/M252Y/V308P/P329K/N434Y


F661
3.40E−08
S239K/M252Y/V308P/P329R/N434Y


F663
6.40E−09
S239K/M252Y/N286E/T307Q/V308P/Q311A/N434Y


F664
3.90E−08
M252Y/N286A/V308P/N434Y


F665
2.00E−08
M252Y/N286D/V308P/N434Y


F666
2.10E−08
M252Y/N286F/V308P/N434Y


F667
3.00E−08
M252Y/N286G/V308P/N434Y


F668
4.00E−08
M252Y/N286H/V308P/N434Y


F669
3.50E−08
M252Y/N286I/V308P/N434Y


F670
2.10E−07
M252Y/N286K/V308P/N434Y


F671
2.20E−08
M252Y/N286L/V308P/N434Y


F672
2.40E−08
M252Y/N286M/V308P/N434Y


F673
2.30E−08
M252Y/N286P/V308P/N434Y









Table 5-17 is a continuation table of Table 5-16.











TABLE 5-17







F674
3.20E−08
M252Y/N286Q/V308P/N434Y


F675
5.10E−08
M252Y/N286R/V308P/N434Y


F676
3.20E−08
M252Y/N286S/V308P/N434Y


F677
4.70E−08
M252Y/N286T/V308P/N434Y


F678
3.30E−08
M252Y/N286V/V308P/N434Y


F679
1.70E−08
M252Y/N286W/V308P/N434Y


F680
1.50E−08
M252Y/N286Y/V308P/N434Y


F681
4.90E−08
M252Y/K288A/V308P/N434Y


F682
8.20E−08
M252Y/K288D/V308P/N434Y


F683
5.00E−08
M252Y/K288E/V308P/N434Y


F684
5.10E−08
M252Y/K288F/V308P/N434Y


F685
5.30E−08
M252Y/K288G/V308P/N434Y


F686
4.60E−08
M252Y/K288H/V308P/N434Y


F687
4.90E−08
M252Y/K288I/V308P/N434Y


F688
2.80E−08
M252Y/K288L/V308P/N434Y


F689
4.10E−08
M252Y/K288M/V308P/N434Y


F690
1.00E−07
M252Y/K288N/V308P/N434Y


F691
3.20E−07
M252Y/K288P/V308P/N434Y


F692
3.90E−08
M252Y/K288Q/V308P/N434Y


F693
3.60E−08
M252Y/K288R/V308P/N434Y


F694
4.70E−08
M252Y/K288V/V308P/N434Y


F695
4.00E−08
M252Y/K288W/V308P/N434Y


F696
4.40E−08
M252Y/K288Y/V308P/N434Y


F697
3.10E−08
S239K/M252Y/V308P/N325G/N434Y


F698
2.20E−08
M252Y/N286E/T307Q/Q311A/N434Y


F699
2.30E−08
S239K/M252Y/N286E/T307Q/Q311A/N434Y


F700
5.20E−08
M252Y/V308P/L328E/N434Y


F705
7.10E−09
M252Y/N286E/V308P/M428I/N434Y


F706
1.80E−08
M252Y/N286E/T307Q/Q311A/M428I/N434Y


F707
5.90E−09
M252Y/N286E/T307Q/V308P/Q311A/N434Y


F708
4.10E−09
M252Y/N286E/T307Q/V308P/Q311A/M428I/N434Y


F709
2.00E−08
S239K/M252Y/N286E/T307Q/Q311A/M428I/N434Y


F710
1.50E−08
P238D/M252Y/N286E/T307Q/Q311A/M428I/N434Y


F711
6.50E−08
S239K/M252Y/T307Q/Q311A/N434Y









Table 5-18 is a continuation table of Table 5-17.











TABLE 5-18







F712
6.00E−08
P238D/M252Y/T307Q/Q311A/N434Y


F713
2.00E−08
P238D/M252Y/N286E/T307Q/Q311A/N434Y


F714
2.30E−07
P238D/M252Y/N325S/N434Y


F715
2.30E−07
P238D/M252Y/N325M/N434Y


F716
2.70E−07
P238D/M252Y/N325L/N434Y


F717
2.60E−07
P238D/M252Y/N325I/N434Y


F718
2.80E−07
P238D/M252Y/Q295M/N434Y


F719
7.40E−08
P238D/M252Y/N325G/N434Y


F720
2.40E−08
M252Y/T307Q/V308P/Q311A/N434Y


F721
1.50E−08
M252Y/T307Q/V308P/Q311A/M428I/N434Y


F722
2.70E−07
P238D/M252Y/A327G/N434Y


F723
2.80E−07
P238D/M252Y/L328D/N434Y


F724
2.50E−07
P238D/M252Y/L328E/N434Y


F725
4.20E−08
L235K/G237R/S239K/M252Y/V308P/N434Y


F726
3.70E−08
L235K/P238K/S239K/M252Y/V308P/N434Y


F729
9.20E−07
T307A/Q311A/N434Y


F730
6.00E−07
T307Q/Q311A/N434Y


F731
8.50E−07
T307A/Q311H/N434Y


F732
6.80E−07
T307Q/Q311H/N434Y


F733
3.20E−07
M252Y/L328E/N434Y


F734
3.10E−07
G236D/M252Y/L328E/N434Y


F736
3.10E−07
M252Y/S267M/L328E/N434Y


F737
3.10E−07
M252Y/S267L/L328E/N434Y


F738
3.50E−07
P238D/M252Y/T307P/N434Y


F739
2.20E−07
M252Y/T307P/Q311A/N434Y


F740
2.90E−07
M252Y/T307P/Q311H/N434Y


F741
3.10E−07
P238D/T250A/M252Y/N434Y


F744
9.90E−07
P238D/T250F/M252Y/N434Y


F745
6.60E−07
P238D/T250G/M252Y/N434Y


F746
6.00E−07
P238D/T250H/M252Y/N434Y


F747
2.80E−07
P238D/T250I/M252Y/N434Y


F749
5.10E−07
P238D/T250L/M252Y/N434Y


F750
3.00E−07
P238D/T250M/M252Y/N434Y


F751
5.30E−07
P238D/T250N/M252Y/N434Y









Table 5-19 is a continuation table of Table 5-18.











TABLE 5-19







F753
1.80E−07
P238D/T250Q/M252Y/N434Y


F755
3.50E−07
P238D/T250S/M252Y/N434Y


F756
3.70E−07
P238D/T250V/M252Y/N434Y


F757
1.20E−06
P238D/T250W/M252Y/N434Y


F758
1.40E−06
P238D/T250Y/M252Y/N434Y


F759

L235K/S239K


F760

L235R/S239K


F761
1.10E−06
P238D/N434Y


F762
3.60E−08
L235K/S239K/M252Y/N286E/T307Q/Q311A/N434Y


F763
3.50E−08
L235R/S239K/M252Y/N286E/T307Q/Q311A/N434Y


F764
6.30E−07
P238D/T307Q/Q311A/N434Y


F765
8.50E−08
P238D/M252Y/T307Q/L309E/Q311A/N434Y


F766
6.00E−07
T307A/L309E/Q311A/N434Y


F767
4.30E−07
T307Q/L309E/Q311A/N434Y


F768
6.40E−07
T307A/L309E/Q311H/N434Y


F769
4.60E−07
T307Q/L309E/Q311H/N434Y


F770
3.00E−07
M252Y/T256A/N434Y


F771
4.00E−07
M252Y/E272A/N434Y


F772
3.80E−07
M252Y/K274A/N434Y


F773
3.90E−07
M252Y/V282A/N434Y


F774
4.00E−07
M252Y/N286A/N434Y


F775
6.20E−07
M252Y/K338A/N434Y


F776
3.90E−07
M252Y/K340A/N434Y


F777
3.90E−07
M252Y/E345A/N434Y


F779
3.90E−07
M252Y/N361A/N434Y


F780
3.90E−07
M252Y/Q362A/N434Y


F781
3.70E−07
M252Y/S375A/N434Y


F782
3.50E−07
M252Y/Y391A/N434Y


F783
4.00E−07
M252Y/D413A/N434Y


F784
5.00E−07
M252Y/L309A/N434Y


F785
7.40E−07
M252Y/L309H/N434Y


F786
2.80E−08
M252Y/S254T/N286E/T307Q/Q311A/N434Y


F787
8.80E−08
M252Y/S254T/T307Q/L309E/Q311A/N434Y


F788
4.10E−07
M252Y/N315A/N434Y









Table 5-20 is a continuation table of Table 5-19.











TABLE 5-20







F789
1.50E−07
M252Y/N315D/N434Y


F790
2.70E−07
M252Y/N315E/N434Y


F791
4.40E−07
M252Y/N315F/N434Y


F792
4.40E−07
M252Y/N315G/N434Y


F793
3.30E−07
M252Y/N315I/N434Y


F791
4.10E−07
M252Y/N315K/N434Y


F795
3.10E−07
M252Y/N315L/N434Y


F796
3.40E−07
M252Y/N315M/N434Y


F798
3.50E−07
M252Y/N315Q/N434Y


F799
4.10E−07
M252Y/N315R/N434Y


F800
3.80E−07
M252Y/N315S/N434Y


F801
4.40E−07
M252Y/N315T/N434Y


F802
3.30E−07
M252Y/N315V/N434Y


F803
3.60E−07
M252Y/N315W/N434Y


F804
4.00E−07
M252Y/N315Y/N434Y


F805
3.00E−07
M252Y/N325A/N434Y


F806
3.10E−07
M252Y/N384A/N434Y


F807
3.20E−07
M252Y/N389A/N434Y


F808
3.20E−07
M252Y/N389A/N390A/N434Y


F809
2.20E−07
M252Y/S254T/T256S/N434Y


F810
2.20E−07
M252Y/A378V/N434Y


F811
4.90E−07
M252Y/E380S/N434Y


F812
2.70E−07
M252Y/E382V/N434Y


F813
2.80E−07
M252Y/S424E/M434Y


F814
1.20E−07
M252Y/N434Y/Y436I


F815
5.50E−07
M252Y/N434Y/T437R


F816
3.60E−07
P238D/T250V/M252Y/T307P/N434Y


F817
9.80E−08
P238D/T250V/M252Y/T307Q/Q311A/N434Y


F819
1.40E−07
P238D/M252Y/N286E/N434Y


F820
3.40E−07
L235K/S239K/M252Y/N434Y


F821
3.10E−07
L235R/S239K/M252Y/N434Y


F822
1.10E−06
P238D/T250Y/M252Y/W313Y/N434Y


F823
1.10E−06
P238D/T250Y/M252Y/W313F/N434Y


F828
2.50E−06
P238D/T250V/M252Y/I253V/N434Y









Table 5-21 is a continuation table of Table 5-20.











TABLE 5-21







F831
1.60E−06
P238D/T250V/M252Y/R255A/N434Y


F832
2.60E−06
P238D/T250V/M252Y/R255D/N434Y


F833
8.00E−07
P238D/T250V/M252Y/R255E/N434Y


F834
8.10E−07
P238D/T250V/M252Y/R255F/N434Y


F836
5.00E−07
P238D/T250V/M252Y/R255H/N434Y


F837
5.60E−07
P238D/T250V/M252Y/R255I/N434Y


F838
4.30E−07
P238D/T250V/M252Y/R255K/N434Y


F839
3.40E−07
P238D/T250V/M252Y/R255L/N434Y


F840
4.20E−07
P238D/T250V/M252Y/R255M/N434Y


F841
1.10E−06
P238D/T250V/M252Y/R255N/N434Y


F843
6.60E−07
P238D/T250V/M252Y/R255Q/N434Y


F844
1.30E−06
P238D/T250V/M252Y/R255S/N434Y


F847
3.40E−07
P238D/T250V/M252Y/R255W/N434Y


F848
8.30E−07
P238D/T250V/M252Y/R255Y/N434Y


F849
3.30E−07
M252Y/D280A/N434Y


F850
2.90E−07
M252Y/D280E/N434Y


F852
3.30E−07
M252Y/D280G/N434Y


F853
3.20E−07
M252Y/D280H/N434Y


F855
3.20E−07
M252Y/D280K/N434Y


F858
3.20E−07
M252Y/D280N/N434Y


F860
3.30E−07
M252Y/D280Q/N434Y


F861
3.20E−07
M252Y/D280R/N434Y


F862
3.00E−07
M252Y/D280S/N434Y


F863
2.70E−07
M252Y/D280T/N434Y


F867
2.80E−07
M252Y/N384A/N389A/N434Y


F868
2.00E−08
G236A/S239D/M252Y/N286E/T307Q/Q311A/N434Y


F869

G236A/S239D


F870
7.30E−08
L235K/S239K/M252Y/T307Q/Q311A/N434Y


F871
7.10E−08
L235R/S239K/M252Y/T307Q/Q311A/N434Y


F872
1.30E−07
L235K/S239K/M252Y/N286E/N434Y


F873
1.20E−07
L235R/S239K/M252Y/N286E/N434Y


F875
4.80E−07
M252Y/N434Y/Y436A


F877
8.30E−07
M252Y/N434Y/Y436E


F878
1.90E−07
M252Y/N434Y/Y436F









Table 5-22 is a continuation table of Table 5-21.











TABLE 5-22







F879
9.20E−07
M252Y/N434Y/Y436G


F880
3.90E−07
M252Y/N434Y/Y436H


F881
3.10E−07
M252Y/N434Y/Y436K


F882
1.30E−07
M252Y/N434Y/Y436L


F883
2.10E−07
M252Y/N434Y/Y436M


F884
4.00E−07
M252Y/N434Y/Y436N


F888
4.80E−07
M252Y/N434Y/Y436S


F889
2.20E−07
M252Y/N434Y/Y436T


F890
1.10E−07
M252Y/N434Y/Y436V


F891
1.70E−07
M252Y/N434Y/Y436W


F892
7.10E−08
M252Y/S254T/N434Y/Y436I


F893
9.80E−08
L235K/S239K/M252Y/N434Y/Y436I


F894
9.20E−08
L235R/S239K/M252Y/N434Y/Y436I


F895
2.10E−08
L235K/S239K/M252Y/N286E/T307Q/Q311A/N315E/




N434Y


F896
2.00E−08
L235R/S239K/M252Y/N286E/T307Q/Q311A/N315E/




N434Y


F897
9.70E−08
M252Y/N315D/N384A/N389A/N434Y


F898
1.70E−07
M252Y/N315E/N384A/N389A/N434Y


F899
1.10E−07
M252Y/N315D/G316A/N434Y


F900
1.70E−07
M252Y/N315D/G316D/N434Y


F901
1.30E−07
M252Y/N315D/G316E/N434Y


F902
2.20E−07
M252Y/N315D/G316F/N434Y


F903
2.30E−07
M252Y/N315D/G316H/N434Y


F904
1.00E−07
M252Y/N315D/G316I/N434Y


F905
1.30E−07
M252Y/N315D/G316K/N434Y


F906
1.50E−07
M252Y/N315D/G316L/N434Y


F907
1.30E−07
M252Y/N315D/G316M/N434Y


F908
1.50E−07
M252Y/N315D/G316N/N434Y


F909
1.30E−07
M252Y/N315D/G316P/N434Y


F910
1.40E−07
M252Y/N315D/G316Q/N434Y


F911
1.30E−07
M252Y/N315D/G316R/N434Y


F912
1.20E−07
M252Y/N315D/G316S/N434Y


F913
1.10E−07
M252Y/N315D/G316T/N434Y


F914
1.50E−07
M252Y/N315D/G316V/N434Y


F915
2.30E−07
M252Y/N315D/G316W/N434Y









Table 5-23 is a continuation table of Table 5-22.











TABLE 5-23







F917
2.50E−07
M252Y/N286S/N434Y


F918
2.80E−07
M252Y/D280E/N384A/N389A/N434Y


F919
3.30E−07
M252Y/D280G/N384A/N389A/N434Y


F920
2.50E−07
M252Y/N286S/N384A/N389A/N434Y


F921
1.20E−07
M252Y/N286E/N384A/N389A/N434Y


F922
5.90E−08
L235K/S239K/M252Y/N286E/N434Y/Y436I


F923
6.00E−08
L235R/S239K/M252Y/N286E/N434Y/Y436I


F924
3.40E−08
L235K/S239K/M252Y/T307Q/Q311A/N434K/Y436I


F925
3.20E−08
L235R/S239K/M252Y/T307Q/Q311A/N434Y/Y436I


F926
1.10E−07
L235K/S239K/M252Y/S254T/N434Y/Y436I


F927
1.00E−07
L235R/S239K/M252Y/S254T/N434Y/Y436I


F928
2.90E−08
M252Y/T307Q/Q311A/N434Y/Y436I


F929
2.90E−08
M252Y/S254T/T307Q/Q311A/N434Y/Y436I


F930
1.40E−07
P238D/T250V/M252Y/N286E/N434Y


F931
1.20E−07
P238D/T250V/M252Y/N434Y/Y436I


F932
3.20E−07
T250V/M252Y/N434Y


F933
3.00E−07
L234R/P238D/T250V/M252Y/N434Y


F934
3.10E−07
G236K/P238D/T250V/M252Y/N434Y


F935
3.20E−07
G237K/P238D/T250V/M252Y/N434Y


F936
3.20E−07
G237R/P238D/T250V/M252Y/N434Y


F937
3.10E−07
P238D/S239K/T250V/M252Y/N434Y


F938
1.60E−07
L235K/S239K/M252Y/N434Y/Y436V


F939
1.50E−07
L235R/S239K/M252Y/N434Y/Y436V


F940
1.50E−07
P238D/T250V/M252Y/N434Y/Y436V


F941
1.20E−08
M252Y/N286E/T307Q/Q311A/N434Y/Y436V


F942
4.20E−08
L235K/S239K/M252Y/T307Q/Q311A/N434Y/Y436V


F943
4.00E−08
L235R/S239K/M252Y/T307Q/Q311A/N434Y/Y436V


F944
1.70E−07
T250V/M252Y/N434Y/Y436V


F945
1.70E−08
T250V/M252Y/V308P/N434Y/Y436V


F946
4.30E−08
T250V/M252Y/T307Q/Q311A/N434Y/Y436V


F947
1.10E−08
T250V/M252Y/T307Q/V308P/Q311A/N434Y/Y436V


F954
5.30E−07
M252Y/N434Y/H435K/Y436V


F957
7.70E−07
M252Y/N434Y/H435N/Y436V


F960
8.00E−07
M252Y/N434Y/H435R/Y436V









Table 5-24 is a continuation table of Table 5-23.











TABLE 5-24







F966
3.10E−07
M252Y/S254A/N434Y


F970
2.50E−06
M252Y/S254G/N434Y


F971
2.60E−06
M252Y/S254H/N434Y


F972
2.60E−07
M252Y/S254I/N434Y


F978
1.30E−06
M252Y/S254Q/N434Y


F980
1.80E−07
M252Y/S254V/N434Y


F987
4.00E−08
P238D/T250V/M252Y/T307Q/Q311A/N434Y/Y436V


F988
6.90E−08
P238D/T250V/M252Y/N286E/N434Y/Y436V


F989
1.40E−08
L235R/S239K/M252Y/V308P/N434Y/Y436V


F990
9.40E−09
L235R/S239K/M252Y/T307Q/V308P/Q311A/N434Y/Y436V


F991
1.30E−08
L235R/S239K/M252Y/N286E/T307Q/Q311A/N434Y/Y436V


F992
5.10E−08
L235R/S239K/M252Y/T307Q/Q311A/M428I/N434Y/Y436V


F993
3.80E−08
M252Y/T307Q/Q311A/N434Y/Y436V


F994
2.80E−07
M252Y/N325G/N434Y


F995
2.90E−07
L235R/P238D/S239K/M252Y/N434Y


F996
1.30E−07
L235R/P238D/S239K/M252Y/N434Y/Y436V


F997
3.80E−07
K248I/T250V/M252Y/N434Y/Y436V


F998
8.50E−07
K248Y/T250V/M252Y/N434Y/Y436V


F999
2.10E−07
T250V/M252Y/E258H/N434Y/Y436V


F1005

N325G


F1008
1.70E−07
L235R/S239K/T250V/M252Y/N434Y/Y436V


F1009
1.20E−08
L235R/S239K/T250V/M252Y/T307Q/V308P/Q311A/N434Y/Y436V


F1010
1.90E−07
L235R/S239K/M252Y/T307A/Q311H/N434Y


F1011
4.50E−08
T250V/M252Y/V308P/N434Y


F1012
4.70E−08
L235R/S239K/T250V/M252Y/V308P/N434Y


F1013
3.00E−08
T250V/M252Y/T307Q/V308P/Q311A/N434Y


F1014
3.20E−08
L235R/S239K/T250V/M252Y/T307Q/V308P/Q311A/N434Y


F1015
2.20E−08
L235R/S239K/M252Y/T307Q/V308P/Q311A/N434Y


F1016
3.80E−09
T250V/M252Y/N286E/T307Q/V308P/Q311A/N434Y/Y436V


F1017
4.20E−09
L235R/S239K/T250V/M252Y/N286E/T307Q/V308P/Q311A/N431Y/Y436V


F1018
3.20E−09
L235R/S239K/M252Y/N286E/T307Q/V308P/Q311A/N434Y/Y436V


F1019
3.40E−07
P238D/T250V/M252Y/N325G/N434Y


F1020
8.50E−08
P238D/T250V/M252Y/T307Q/Q311A/N325G/N434Y









Table 5-25 is a continuation table of Table 5-24.











TABLE 5-25







F1021
3.30E−07
P238D/T250V/M252Y/N325A/N434Y


F1022

K326D/L328Y


F1023
4.40E−08
S239D/T250V/M252Y/T307Q/Q311A/N434Y/Y436V


F1024
4.00E−08
T250V/M252Y/T307Q/Q311A/K326D/L328Y/N434Y/Y436V


F1025
3.60E−08
S239D/T250V/M252Y/T307Q/Q311A/K326D/L328Y/N434Y/Y436V


F1026
8.40E−08
M252Y/T307A/Q311H/N434Y/Y436V


F1027
8.60E−08
L235R/S239K/M252Y/T307A/Q311H/N434Y/Y436V


F1028
4.60E−08
G236A/S239D/T250V/M252Y/T307Q/Q311A/N434Y/Y436V


F1029
5.10E−08
T250V/M252Y/T307Q/Q311A/I332E/N434Y/Y436V


F1030

I332E


F1031
5.30E−08
G236A/S239D/T250V/M252Y/T307Q/Q311A/I332E/N434Y/Y436V


F1032
4.30E−08
P238D/T250V/M252Y/T307Q/Q311A/N325G/N434Y/Y436V


F1033
1.00E−06
P238D/N434W


F1034
1.50E−08
L235K/S239K/M252Y/V308P/N434Y/Y436V


F1035
1.00E−08
L235K/S239K/M252Y/T307Q/V308P/Q311A/N434Y/Y436V


F1036
1.40E−08
L235K/S239K/M252Y/N286E/T307Q/Q311A/N434Y/Y436V


F1037
6.10E−08
L235K/S239K/M252Y/T307Q/Q311A/M428I/N434Y/Y436V


F1038
2.80E−07
L235K/P238D/S239K/M252Y/N434Y


F1039
1.30E−07
L235K/P238D/S239K/M252Y/N434Y/Y436V


F1040
2.00E−07
L235K/S239K/T250V/M252Y/N434Y/Y436V


F1041
1.40E−08
L235K/S239K/T250V/M252Y/T307Q/V308P/Q311A/N434Y/Y436V


F1042
2.00E−07
L235K/S239K/M252Y/T307A/Q311H/N434Y


F1043
5.20E−08
L235K/S239K/T250V/M252Y/V308P/N434Y


F1044
3.50E−08
L235K/S239K/T250V/M252Y/T307Q/V308P/Q311A/N434Y


F1045
2.50E−08
L235K/S239K/M252Y/T307Q/V308P/Q311A/N434Y


F1046
4.50E−09
L235K/S239K/T250V/M252Y/N286E/T307Q/V308P/Q311A/N434Y/Y436V


F1047
3.40E−09
L235K/S239K/M252Y/N286E/T307Q/V308P/Q311A/N434Y/Y436V


F1048
9.90E−08
L235K/S239K/M252Y/T307A/Q311H/N434Y/Y436V


F1050
3.50E−09
T250V/M252Y/N286E/T307Q/V308P/Q311A/M428I/N434Y/Y436V


F1051
3.90E−09
L235R/S239K/T250V/M252Y/N286E/T307Q/V308P/Q311A/M428I/N434Y/




Y436V


F1052
3.20E−09
L235R/S239K/M252Y/N286E/T307Q/V308P/Q311A/M428I/N434Y/Y436V









Table 5-26 is a continuation table of Table 5-25.











TABLE 5-26







F1053
4.23E−08
L235R/S239K/T250V/M252Y/T307Q/Q311A/




N434Y/Y436V


F1058
1.31E−07
M252Y/Q386E/N434Y/Y436V


F1059
1.39E−07
M252Y/Q386R/N434Y/Y436V


F1060
1.43E−07
M252Y/Q386S/N434Y/Y436V


F1061
1.19E−07
M252Y/P387E/N434Y/Y436V


F1062
 1.2E−07
M252Y/P387R/N434Y/Y436V


F1063
1.43E−07
M252Y/P387S/N434Y/Y436V


F1064
1.32E−07
M252Y/V422E/N434Y/Y436V


F1065
1.38E−07
M252Y/V422R/N434Y/Y436V


F1066
1.45E−07
M252Y/V422S/N434Y/Y436V


F1067
1.26E−07
M252Y/S424E/N434Y/Y436V


F1068
1.69E−07
M252Y/S424R/N434Y/Y436V


F1069
1.39E−07
M252Y/N434Y/Y436V/Q438E


F1070
1.73E−07
M252Y/N434Y/Y436V/Q438R


F1071
1.24E−07
M252Y/N434Y/Y436V/Q438S


F1072
1.35E−07
M252Y/N434Y/Y436V/S440E


F1073
1.34E−07
M252Y/N434Y/Y436V/S440R


F1074
1.32E−07
S239D/M252Y/N434Y/Y436V


F1075
 1.4E−07
M252Y/K326D/L328Y/N434Y/Y436V


F1076
1.27E−07
S239D/M252Y/K326D/L328Y/N434Y/Y436V


F1077
2.03E−06
K248N/M252Y/N434Y


F1078
 4.7E−07
M252Y/E380N/E382S/N434Y


F1079
3.44E−07
M252Y/E382N/N384S/N434Y


F1080
3.19E−07
M252Y/S424N/N434Y


F1081
 6.2E−07
M252Y/N434Y/Y436N/Q438T


F1082
2.76E−07
M252Y/N434Y/Q438N


F1083
3.45E−07
M252Y/N434Y/S440N


F1094
 2.6E−07
M252Y/N434Y/S442N


F1095
2.86E−07
M252Y/S383N/G385S/N434Y


F1096
2.72E−07
M252Y/Q386T/N434Y


F1097
2.82E−07
M252Y/G385N/P387S/N434Y


F1098
2.58E−07
S239D/M252Y/N434Y


F1099
2.57E−07
M252Y/K326D/L328Y/N434Y


F1100
2.41E−07
S239D/M252Y/K326D/L328Y/N434Y


F1101
6.59E−08
S239D/M252Y/T307Q/Q311A/N434Y


F1102
6.46E−08
M252Y/T307Q/Q311A/K326D/L328Y/N434Y


F1103
6.11E−08
S239D/M252Y/T307Q/Q311A/K326D/L328Y/




N434Y


F1104
1.77E−07
M252Y/V422E/S424R/N434Y/Y436V


F1105
1.54E−07
M252Y/V422S/S424R/N434Y/Y436V


F1106
1.42E−07
M252Y/N434Y/Y436V/Q438R/S440E


F1107
1.23E−07
M252Y/V422D/N434Y/Y436V









Table 5-27 is a continuation table of Table 5-26.











TABLE 5-27







F1108
1.26E−07
M252Y/V422K/N434Y/Y436V


F1109
1.27E−07
M252Y/V422T/N434Y/Y436V


F1110
1.33E−07
M252Y/V422Q/N434Y/Y436V


F1111
1.65E−07
M252Y/S424K/N434Y/Y436V


F1112
1.23E−07
M252Y/N434Y/Y436V/Q438K


F1113
1.18E−07
M252Y/N434Y/Y436V/S440D


F1114
1.31E−07
M252Y/N434Y/Y436V/S440Q


F1115
1.35E−07
M252Y/S424N/N434Y/Y436V


F1116
7.44E−08
M252Y/T307Q/Q311A/S424N/N434Y


F1117
4.87E−08
T250V/M252Y/T307Q/Q311A/S424N/N434Y/Y436V


F1118
1.32E−08
T250V/M252Y/T307Q/V308P/Q311A/S424N/N434Y/Y436V


F1119
1.03E−08
T250V/M252Y/T307Q/V308P/Q311A/V422E/N434Y/Y436V


F1120
1.04E−08
T250V/M252Y/T307Q/V308P/Q311A/S424R/N434Y/Y436V


F1121
1.04E−08
T250V/M252Y/T307Q/V308P/Q311A/V422E/S424R/N434Y/Y436V


F1122
1.37E−08
T250V/M252Y/T307Q/V308P/Q311A/N434Y/Y436V/Q438R


F1123
9.55E−09
T250V/M252Y/T307Q/V308P/Q311A/N434Y/Y436V/S440E


F1124
1.22E−08
T250V/M252Y/T307Q/V308P/Q311A/N434Y/Y436V/Q438R/S440E


F1125
5.18E−08
M252V/T307Q/N434Y/Y436V


F1126
8.95E−08
M252Y/T307A/N434Y/Y436V


F1127
7.94E−08
M252Y/Q311A/N434Y/Y436V


F1128
1.17E−07
M252Y/Q311H/N434Y/Y436V


F1129
4.48E−08
M252Y/T307Q/Q311H/N434Y/Y436V


F1130
5.54E−08
M252Y/T307A/Q311A/N434Y/Y436V


F1131
1.29E−07
L235R/S239K/M252Y/V422E/N434Y/Y436V


F1132
 1.4E−07
L235R/S239K/M252Y/V422S/N434Y/Y436V


F1133
1.58E−07
L235R/S239K/M252Y/S424R/N434Y/Y436V


F1134
1.66E−07
L235R/S239K/M252Y/N434Y/Y436V/Q438R


F1135
1.26E−07
L235R/S239K/M252Y/N434Y/Y436V/S440E


F1136
1.63E−07
L235R/S239K/M252Y/V422E/S424R/N434Y/Y436V


F1137
1.58E−07
L235R/S239K/M252Y/V422S/S424R/N434Y/Y436V


F1138
1.65E−07
L235R/S239K/M252Y/N434Y/Y436V/Q438R/S440E


F1139
1.52E−07
L235R/S239K/M252Y/S424N/N434Y/Y436V


F1140
1.62E−07
M252Y/V422E/S424R/N434Y/Y436V/Q438R/S440E


F1141
1.77E−07
M252Y/V422S/S424R/N434Y/Y436V/Q438R/S440E


F1142
1.87E−07
L235R/S239K/M252Y/V422E/S424R/N434Y/Y436V/Q438R/S440E


F1143
1.98E−07
L235R/S239K/M252Y/V422S/S424R/N434Y/Y436V/Q438R/S440E


F1144
1.44E−08
L235R/S239K/T250V/M252Y/T307Q/V308P/Q311A/N434Y/Y436V/Q438R/S440E


F1145
5.23E−08
T250V/M252Y/T307Q/Q311A/N434Y/Y436V/Q438R/S440E


F1146
6.24E−08
L235R/S239K/T250V/M252Y/T307Q/Q311A/N434Y/Y436V/Q438R/S440E


F1147
7.19E−08
M252Y/T307Q/Q311A/N434Y/Q438R/S440E









Table 5-28 is a continuation table of Table 5-27.











TABLE 5-28







F1148
7.63E−08
L235R/S239K/M252Y/T307Q/Q311A/N434Y/Q438R/S440E


F1151
2.51E−07
L235R/S239K/M252Y/S424N/N434Y


F1152
7.38E−08
L235R/S239K/M252Y/T307Q/Q311A/S424N/N434Y


F1153
4.85E−08
L235R/S239K/T250V/M252Y/T307Q/Q311A/S424N/N434Y/Y436V


F1154
1.34E−08
L235R/S239K/T250V/M252Y/T307Q/V308P/Q311A/S424N/N434Y/Y436V


F1157
2.09E−07
M252Y/N434Y/Q438R/S440E


F1158
2.44E−07
L235R/S239K/M252Y/N434Y/Q438R/S440E


F1159
4.79E−07
S424N/N434W


F1160
2.88E−07
V308F/S424N/N434Y


F1161
1.07E−06
I332V/S424N/N434Y


F1162
3.43E−07
P238D/T250Y/M252Y/N434Y/Y436V


F1163
1.54E−07
P238D/T250Y/M252Y/T307Q/Q311A/N434Y


F1164
6.96E−08
P238D/T250Y/M252Y/T307Q/Q311A/N434Y/Y436V


F1165
1.63E−08
P238D/T250Y/M252Y/T307Q/V308P/Q311A/N434Y/Y436V


F1174
 4.9E−07
P257I/N434H


F1176
1.98E−06
V308F


F1178
8.72E−07
V259I/V308F/M428L


F1183
1.28E−06
E380A/M428L/N434S


F1184
  1E−06
T307A/M428L/N434S


F1185
9.17E−07
T307A/E380A/M428L/N434S


F1188
1.72E−06
T307A/E380A/N434H


F1189
1.57E−07
M252Y/H433D/N434Y/Y436V/Q438R/S440E


F1190
 2.4E−07
M252Y/H433E/N434Y/Y436V/Q438R/S440E


F1191
2.11E−07
M252Y/N434Y/Y436V/T437A/Q438R/S440E


F1192
1.27E−07
M252Y/N434Y/Y436V/T437G/Q438R/S440E


F1194
1.55E−07
M252Y/N434Y/Y436V/Q438R/K439D/S440E


F1195
1.76E−07
M252Y/N434Y/Y436V/Q438R/S440E/L441A


F1196
1.51E−07
M252Y/N434Y/Y436V/Q438R/S440E/L441E


F1197
9.46E−08
M252Y/S254T/N434Y/Y436V/Q438R/S440E


F1198
7.83E−08
M252Y/T256E/N434Y/Y436V/Q438R/S440E


F1199
6.25E−08
M252Y/S254T/T256E/N434Y/Y436V/Q438R/S440E


F1200
1.26E−07
T250V/M252Y/S254T/N434Y/Y436V/Q438R/S440E


F1201
1.07E−07
T250V/M252Y/T256E/N434Y/Y436V/Q438R/S440E


F1202
8.81E−08
T250V/M252Y/S254T/T256E/N434Y/Y436V/Q438R/S440E


F1203
1.52E−07
M252Y/T256Q/N434Y/Y436V/Q438R/S440E


F1204
1.18E−07
M252Y/S254T/T256Q/N434Y/Y436V/Q438R/S440E


F1205
1.98E−07
T250V/M252Y/T256Q/N434Y/Y436V/Q438R/S440E


F1206
1.69E−07
T250V/M252Y/S254T/T256Q/N434Y/Y436V/Q438R/S440E


F1207
1.11E−06
I332E/M428L/N434S


F1208
5.71E−07
L251A/M252Y/N434Y/Y436V


F1211
1.23E−06
L251H/M252Y/N434Y/Y436V









Table 5-29 is a continuation table of Table 5-28.











TABLE 5-29







F1213
6.33E−07
L251N/M252Y/N434Y/Y436V


F1216
1.16E−06
L251S/M252Y/N434Y/Y436V


F1217
1.14E−06
L251T/M252Y/N434Y/Y436V


F1218
2.51E−07
L251V/M252Y/N434Y/Y436V


F1229
2.81E−06
M252Y/I253V/N434Y/Y436V


F1230
1.12E−07
M252Y/N434Y/Y436V/Q438R/S440D


F1231
9.73E−08
M252Y/N434Y/Y436V/Q438K/S440E


F1232
9.79E−08
M252Y/N434Y/Y436V/Q438K/S440D


F1243
1.25E−07
L235R/S239K/M252Y/S254T/N434Y/Y436V/Q438R/S440E


F1244
1.02E−07
L235R/S239K/M252Y/T256E/N434Y/Y436V/Q438R/S440E


F1245
 8.2E−08
L235R/S239K/M252Y/S254T/T256E/N434Y/Y436V/Q438R/S440E


F1246
1.73E−07
L235R/S239K/T250V/M252Y/S254T/N434Y/Y436V/Q438R/S440E


F1247
1.45E−07
L235R/S239K/T250V/M252Y/T256E/N434Y/Y436V/Q438R/S440E


F1248
 1.2E−07
L235R/S239K/T250V/M252Y/S254T/T256E/N434Y/Y436V/Q438R/S440E


F1249
2.06E−07
L235R/S239K/M252Y/T256Q/N434Y/Y436V/Q438R/S440E


F1250
1.66E−07
L235R/S239K/M252Y/S254T/T256Q/N434Y/Y436V/Q438R/S440E


F1251
2.77E−07
L235R/S239K/T250V/M252Y/T256Q/N434Y/Y436V/Q438R/S440E


F1252
2.33E−07
L235R/S239K/T250V/M252Y/S254T/T256Q/N434Y/Y436V/Q438R/S440E


F1253
1.12E−07
L235R/S239K/M252Y/T307A/N434Y/Y436V/Q438R/S440E


F1254
6.42E−08
L235R/S239K/M252Y/T307Q/N434Y/Y436V/Q438R/S440E


F1255
1.11E−07
L235R/S239K/M252Y/Q311A/N434Y/Y436V/Q438R/S440E


F1256
1.56E−07
L235R/S239K/M252Y/Q311H/N434Y/Y436V/Q438R/S440E


F1257
7.81E−08
L235R/S239K/M252Y/T307A/Q311A/N434Y/Y436V/Q438R/S440E


F1258
1.05E−07
L235R/S239K/M252Y/T307A/Q311H/N434Y/Y436V/Q438R/S440E


F1259
4.46E−08
L235R/S239K/M252Y/T307Q/Q311A/N434Y/Y436V/Q438R/S440E


F1260
6.53E−08
L235R/S239K/M252Y/T307Q/Q311H/N434Y/Y436V/Q438R/S440E


F1261
1.35E−07
L235R/S239K/M252Y/N434Y/Y436V/Q438R/S440D


F1262
1.26E−07
L235R/S239K/M252Y/N434Y/Y436V/Q438K/S440E


F1263
1.24E−07
L235R/S239K/M252Y/N434Y/Y436V/Q438K/S440D


F1264
1.27E−07
L235R/S239K/M252Y/T256A/N434Y/Y436V/Q438R/S440E


F1265
1.57E−07
L235R/S239K/M252Y/T256G/N434Y/Y436V/Q438R/S440E


F1266
9.99E−08
L235R/S239K/M252Y/T256N/N434Y/Y436V/Q438R/S440E


F1267
 1.5E−07
L235R/S239K/M252Y/S254A/N434Y/Y436V/Q438R/S440E


F1268
  2E−07
L235R/S239K/M252Y/H433D/N434Y/Y436V/Q438R/S440E


F1269
1.69E−07
L235R/S239K/M252Y/H433D/N434Y/Y436V/Q438K/S440D


F1270
1.18E−07
L235R/S239K/M252Y/S254A/N434Y/Y436V/Q438K/S440D


F1271
2.05E−07
L235R/S239K/M252Y/S254A/H433D/N434Y/Y436V/Q438R/S440E


F1272
1.71E−07
L235R/S239K/M252Y/S254A/H433D/N434Y/Y436V/Q438K/S440D


F1273
1.53E−07
L235R/S239K/M252Y/T256Q/N434Y/Y436V/Q438K/S440D


F1274
2.48E−07
L235R/S239K/M252Y/T256Q/H433D/N434Y/Y436V/Q438R/S440E


F1275
2.09E−07
L235R/S239K/M252Y/T256Q/H433D/N434Y/Y436V/Q438K/S440D









Table 5-30 is a continuation table of Table 5-29.











TABLE 5-30







F1276
1.02E−07
L235R/S239K/M252Y/T256A/N434Y/Y436V/Q438K/S440D


F1277
1.69E−07
L235R/S239K/M252Y/T256A/H433D/N434Y/Y436V/Q438R/S440E


F1278
 1.4E−07
L235R/S239K/M252Y/T256A/H433D/N434Y/Y436V/Q438K/S440D


F1279
1.23E−07
L235R/S239K/M252Y/T256G/N434Y/Y436V/Q438K/S440D


F1280
2.09E−07
L235R/S239K/M252Y/T256G/H433D/N434Y/Y436V/Q438R/S440E


F1281
1.74E−07
L235R/S239K/M252Y/T256G/H433D/N434Y/Y436V/Q438K/S440D


F1282
7.69E−08
L235R/S239K/M252Y/T256N/N434Y/Y436V/Q438K/S440D


F1283
1.34E−07
L235R/S239K/M252Y/T256N/H433D/N434Y/Y436V/Q438R/S440E


F1284
1.12E−07
L235R/S239K/M252Y/T256N/H433D/N434Y/Y436V/Q438K/S440D


F1285
9.36E−08
L235R/S239K/M252Y/S254T/N434Y/Y436V/Q438K/S440D


F1286
1.57E−07
L235R/S239K/M252Y/S254T/H433D/N434Y/Y436V/Q438R/S440E


F1287
 1.5E−07
L235R/S239K/M252Y/S254T/H433D/N434Y/Y436V/Q438K/S440D


F1288
7.95E−08
L235R/S239K/M252Y/T256E/N434Y/Y436V/Q438K/S440D


F1289
1.33E−07
L235R/S239K/M252Y/T256E/H433D/N434Y/Y436V/Q438R/S440E


F1290
1.11E−07
L235R/S239K/M252Y/T256E/H433D/N434Y/Y436V/Q438K/S440D


F1291
1.51E−07
L235R/S239K/M252Y/H433D/N434Y/Y436V


F1292
4.24E−07
L235R/S239K/H433D/N434W/Y436V/Q438R/S440E


F1293
1.61E−07
L235R/S239K/M252Y/T256E/N434Y/Q438R/S440E


F1294
  2E−07
L235R/S239K/M252Y/T256E/N434Y/Y436T/Q438R/S440E


F1295
9.84E−08
L235R/S239K/M252Y/T256E/N434Y/Y436F/Q438R/S440E


F1296
2.27E−07
L235R/S239K/M252Y/T256E/H433D/N434Y/Q438R/S440E


F1297
 2.5E−07
L235R/S239K/M252Y/T256E/H433D/N434Y/Y436T/Q438R/S440E


F1298
1.47E−07
L235R/S239K/M252Y/T256E/H433D/N434Y/Y436F/Q438R/S440E


F1299
 1.5E−07
L235R/S239K/M252Y/T256E/N434Y/Q438K/S440D


F1300
1.63E−07
L235R/S239K/M252Y/T256E/N434Y/Y436T/Q438K/S440D


F1301
 8.3E−08
L235R/S239K/M252Y/T256E/N434Y/Y436F/Q438K/S440D


F1302
2.15E−07
L235R/S239K/M252Y/T256E/H433D/N434Y/Q438K/S440D


F1303
 2.1E−07
L235R/S239K/M252Y/T256E/H433D/N434Y/Y436T/Q438K/S440D


F1304
1.24E−07
L235R/S239K/M252Y/T256E/H433D/N434Y/Y436F/Q438K/S440D


F1305
2.05E−07
L235R/S239K/M252Y/H433D/N434Y/Y436V/Q438R/S440D


F1306
1.92E−07
L235R/S239K/M252Y/H433D/N434Y/Y436V/Q438K/S440E


F1307
1.44E−07
L235R/S239K/M252Y/V422A/S424A/N434Y/Y436V


F1308
2.06E−07
L235R/S239K/M252Y/V422L/S424L/N434Y/Y436V


F1309
1.26E−07
L235R/S239K/M252Y/N434Y/Y436V/Q438A/S440A


F1310
2.28E−07
L235R/S239K/M252Y/N434Y/Y436V/Q438L/S440L


F1311
1.69E−07
L235R/S239K/M252Y/V422A/S424A/H433D/N434Y/Y436V


F1312
1.79E−07
L235R/S239K/M252Y/V422L/S424L/H433D/N434Y/Y436V


F1313
1.77E−07
L235R/S239K/M252Y/H433D/N434Y/Y436V/Q438A/S440A


F1314
2.27E−07
L235R/S239K/M252Y/H433D/N434Y/Y436V/Q438L/S440L


F1315
1.52E−07
G237K/S239K/M252Y/N434Y/Y436V


F1316
1.49E−07
G237R/S239K/M252Y/N434Y/Y436V









Table 5-31 is a continuation table of Table 5-30.











TABLE 5-31







F1317
1.38E−07
S239K/M252Y/P329K/N434Y/Y436V


F1318
1.43E−07
S239K/M252Y/P329R/N434Y/Y436V


F1319
2.67E−07
M252Y/L328Y/N434Y


F1320
1.22E−07
L235R/S239K/M252Y/S254T/N434Y/Y436V/Q438R/S440D


F1321
1.03E−07
L235R/S239K/M252Y/S254T/N434Y/Y436V/Q438K/S440E


F1322
 1.6E−07
L235R/S239K/M252Y/S254T/H433D/N434Y/Y436V/Q438R/S440D


F1323
1.49E−07
L235R/S239K/M252Y/S254T/H433D/N434Y/Y436V/Q438K/S440E


F1324
1.32E−07
L234A/L235A/M252Y/N434Y/Y436V


F1325
2.13E−07
L234A/L235A/M252Y/N297A/N434Y/Y436V


F1326
1.09E−08
L234A/L235A/T250V/M252Y/T307Q/V308P/Q311A/N434Y/Y436V


F1327
1.41E−08
L234A/L235A/T250V/M252Y/N297A/T307Q/V308P/Q311A/N434Y/Y436V


F1328
1.52E−07
L235R/G236R/S239K/M252Y/N434Y/Y436V/Q438R/S440E


F1329
1.29E−07
L235R/G236R/S239K/M252Y/S254T/N434Y/Y436V/Q438R/S440E


F1330
1.03E−07
L235R/G236R/S239K/M252Y/T256E/N434Y/Y436V/Q438R/S440E


F1331
7.75E−08
L235R/G236R/S239K/M252Y/S254T/T256E/N434Y/Y436V/Q438R/S440E


F1333
1.23E−07
L235R/G236R/S239K/M252Y/N434Y/Y436V


F1334
1.04E−07
L235R/G236R/S239K/M252Y/N434Y/Y436V/Q438K/S440D


F1335
8.78E−08
L235R/G236R/S239K/M252Y/S254T/N434Y/Y436V/Q438K/S440D


F1336
7.18E−08
L235R/G236R/S239K/M252Y/T256E/N434Y/Y436V/Q438K/S440D


F1337
7.41E−08
L235R/S239K/M252Y/T256E/N434Y/Y436V/Q438K/S440E


F1338
1.04E−07
L235R/S239K/M252Y/T256E/H433D/N434Y/Y436V/Q438K/S440E


F1339
2.51E−07
L235R/S239K/M252Y/S254T/T256E/H433D/N434Y/Y436T/Q438K/S440E


F1340
5.58E−08
L235R/S239K/M252Y/S254T/T256E/N434Y/Y436V/Q438K/S440E


F1341
3.22E−07
L235R/S239K/M252Y/S254T/N434Y/Y436T/Q438K/S440E


F1342
2.51E−07
L235R/S239K/M252Y/T256E/N434Y/Y436T/Q438K/S440E


F1343
2.01E−07
L235R/S239K/M252Y/S254T/T256E/N434Y/Y436T/Q438K/S440E


F1344
3.96E−07
L235R/S239K/M252Y/N434Y/Y436T/Q438K/S440E


F1345
1.05E−07
L235R/G236R/S239K/M252Y/N434Y/Y436V/Q438K/S440E


F1346
8.59E−08
L235R/G236R/S239K/M252Y/S254T/N434Y/Y436V/Q438K/S440E


F1347
7.14E−08
L235R/G236R/S239K/M252Y/T256E/N434Y/Y436V/Q438K/S440E


F1348
5.52E−08
L235R/G236R/S239K/M252Y/S254T/T256E/N434Y/Y436V/Q438K/S440E


F1349
3.36E−07
L235R/S239K/M252Y/N434Y/Y436T/Q438R/S440E


F1350
1.18E−07
L235R/S239K/M252Y/N434Y/Y436F/Q438K/S440E


F1351
1.62E−07
L235R/S239K/M252Y/N434Y/Y436F/Q438R/S440E


F1352
3.93E−07
L235R/S239K/M252Y/H433D/N434Y/Y436T/Q438K/S440E


F1353
4.33E−07
L235R/S239K/M252Y/H433D/N434Y/Y436T/Q438R/S440E


F1354
2.29E−07
L235R/S239K/M252Y/H433D/N434Y/Y436F/Q438K/S440E


F1355
2.47E−07
L235R/S239K/M252Y/H433D/N434Y/Y436F/Q438R/S440E


F1356
1.58E−07
G236R/M252Y/L328R/N434Y/Y436V


F1357
2.81E−07
L235R/S239K/M252Y/S254T/N434Y/Y436T/Q438R/S440E


F1358
9.07E−08
L235R/S239K/M252Y/S254T/N434Y/Y436F/Q438K/S440E









Table 5-32 is a continuation table of Table 5-31.











TABLE 5-32







F1359
1.28E−07
L235R/S239K/M252Y/S254T/N434Y/Y436F/Q438R/S440E


F1360
3.12E−07
L235R/S239K/M252Y/S254T/H433D/N434Y/Y436T/Q438K/S440E


F1361
3.52E−07
L235R/S239K/M252Y/S254T/H433D/N434Y/Y436T/Q438R/S440E


F1362
1.41E−07
L235R/S239K/M252Y/S254T/H433D/N434Y/Y436F/Q438K/S440E


F1363
 1.9E−07
L235R/S239K/M252Y/S254T/H433D/N434Y/Y436F/Q438R/S440E


F1364
7.49E−08
L235R/S239K/M252Y/T256E/N434Y/Y436F/Q438K/S440E


F1365
3.14E−07
L235R/S239K/M252Y/T256E/H433D/N434Y/Y436T/Q438K/S440E


F1366
1.17E−07
L235R/S239K/M252Y/T256E/H433D/N434Y/Y436F/Q438K/S440E


F1367
1.79E−07
L235R/S239K/M252Y/S254T/T256E/N434Y/Y436T/Q438R/S440E


F1368
5.49E−08
L235R/S239K/M252Y/S254T/T256E/N434Y/Y436F/Q438K/S440E


F1369
 7.6E−08
L235R/S239K/M252Y/S254T/T256E/N434Y/Y436F/Q438R/S440E


F1370
9.14E−08
L235R/S239K/M252Y/S254T/T256E/H433D/N434Y/Y436V/Q438K/S440E


F1371
1.09E−07
L235R/S239K/M252Y/S254T/T256E/H433D/N434Y/Y436V/Q438R/S440E


F1372
2.28E−07
L235R/S239K/M252Y/S254T/T256E/H433D/N434Y/Y436T/Q438R/S440E


F1373
8.67E−08
L235R/S239K/M252Y/S254T/T256E/H433D/N434Y/Y436F/Q438K/S440E


F1374
 1.2E−07
L235R/S239K/M252Y/S254T/T256E/H433D/N434Y/Y436F/Q438R/S440E


F1375
1.03E−07
L235R/S239K/M252Y/S254T/N434Y/Y436V


F1376
9.09E−08
L235R/S239K/M252Y/S254T/T256E/N434Y/Y436V


F1377
8.27E−08
L235R/S239K/M252Y/T256E/N434Y/Y436V


F1378
3.61E−07
L235R/S239K/M252Y/N434Y/Y436T


F1379
2.85E−07
L235R/S239K/M252Y/N434Y/Y436F









Fcγ Receptor


Fcγ receptor (also described as FcγR) refers to a receptor capable of binding to the Fc region of monoclonal IgG1, IgG2, IgG3, or IgG4 antibodies, and includes all members belonging to the family of proteins substantially encoded by an Fcγ receptor gene. In human, the family includes FcγRI (CD64) including isoforms FcγRIa, FcγRIb and FcγRIc; FcγRII (CD32) including isoforms FcγRIIa (including allotypes H131 and R131, i.e., FcγRIIa(H) and FcγRIIa(R)), FcγRIIb (including FcγRIIb-1 and FcγRIIb-2), and FcγRIIc; and FcγRIII (CD16) including isoforms FcγRIIIa (including allotypes V158 and F158, i.e., FcγRIIIa(V) and FcγRIIIa(F)) and FcγRIIIb (including allotypes FcγRIIIb-NA1 and FcγRIIIb-NA2); as well as all unidentified human FcγRs, FcγR isoforms, and allotypes thereof. However, Fcγ receptor is not limited to these examples. Without being limited thereto, FcγR includes those derived from humans, mice, rats, rabbits, and monkeys. FcγR may be derived from any organisms. Mouse FcγR includes, without being limited to, FcγRI (CD64), FcγRII (CD32), FcγRIII (CD16), and FcγRIII-2 (FcγRIV, CD16-2), as well as all unidentified mouse FcγRs, FcγR isoforms, and allotypes thereof. Such preferred Fcγ receptors include, for example, human FcγRI (CD64), FcγRIIa (CD32), FcγRIIb (CD32), FcγRIIIa (CD16), and/or FcγRIIIb (CD16). The polynucleotide sequence and amino acid sequence of FcγRI are shown in SEQ ID NOs: 15 (NM 000566.3) and 16 (NP 000557.1), respectively; the polynucleotide sequence and amino acid sequence of FcγRIIa (allotype H131) are shown in SEQ ID NOs: 17 (BCO20823.1) and 18 (AAH20823.1) (allotype R131 is a sequence in which amino acid at position 166 of SEQ ID NO: 18 is substituted with Arg), respectively; the polynucleotide sequence and amino acid sequence of FcγRIIb are shown in SEQ ID NOs: 19 (BC146678.1) and 20 (AAI46679.1), respectively; the polynucleotide sequence and amino acid sequence of FcγRIIIa are shown in SEQ ID NOs: 21 (BCO33678.1) and 22 (AAH33678.1), respectively; and the polynucleotide sequence and amino acid sequence of FcγRIIIb are shown in SEQ ID NOs: 23 (BC128562.1) and 24 (AAI28563.1), respectively (RefSeq accession number is shown in each parentheses). For example, as described as FcγRIIIaV when allotype V158 is used, unless otherwise specified, allotype F158 is used; however, the allotype of isoform FcγRIIIa described herein should not be interpreted as being particularly limited.


In FcγRI (CD64) including FcγRIa, FcγRIb, and FcγRIc, and FcγRIII (CD16) including isoforms FcγRIIIa (including allotypes V158 and F158) and FcγRIIIb (including allotypes FcγRIIIb-NA1 and FcγRIIIb-NA2), a chain that binds to the Fc portion of IgG is associated with common γ chain having ITAM responsible for transduction of intracellular activation signal. Meanwhile, the cytoplasmic domain of FcγRII (CD32) including isoforms FcγRIIa (including allotypes H131 and R131) and FcγRIIc contains ITAM. These receptors are expressed on many immune cells such as macrophages, mast cells, and antigen-presenting cells. The activation signal transduced upon binding of these receptors to the Fc portion of IgG results in enhancement of the phagocytic activity of macrophages, inflammatory cytokine production, mast cell degranulation, and the enhanced function of antigen-presenting cells. Fcγ receptors having the ability to transduce the activation signal as described above are also referred to as activating Fcγ receptors.


Meanwhile, the cytoplasmic domain of FcγRIIb (including FcγRIIb-1 and FcγRIIb-2) contains ITIM responsible for transduction of inhibitory signals. The crosslinking between FcγRIIb and B cell receptor (BCR) on B cells suppresses the activation signal from BCR, which results in suppression of antibody production via BCR. The crosslinking of FcγRIII and FcγRIIb on macrophages suppresses the phagocytic activity and inflammatory cytokine production. Fcγ receptors having the ability to transduce the inhibitory signal as described above are also referred to as inhibitory Fcγ receptor.


FcγR-Binding Activity of Fc Region

As mentioned above, Fc regions having an Fcγ receptor-binding activity are examples of Fc regions comprised in the antigen-binding molecules of the present invention. A non-limiting embodiment of such an Fc region includes the Fc region of human IgG1 (SEQ ID NO: 9), IgG2 (SEQ ID NO: 10), IgG3 (SEQ ID NO: 11), or IgG4 (SEQ ID NO: 12). Whether an Fcγ receptor has binding activity to the Fc region of a monoclonal IgG1, IgG2, IgG3, or IgG4 antibody can be assessed by ALPHA screen (Amplified Luminescent Proximity Homogeneous Assay), surface plasmon resonance (SPR)-based BIACORE method, and others (Proc. Natl. Acad. Sci. USA (2006) 103(11), 4005-4010), in addition to the above-described FACS and ELISA formats.


ALPHA screen is performed by the ALPHA technology based on the principle described below using two types of beads: donor and acceptor beads. A luminescent signal is detected only when molecules linked to the donor beads interact biologically with molecules linked to the acceptor beads and when the two beads are located in close proximity. Excited by laser beam, the photosensitizer in a donor bead converts oxygen around the bead into excited singlet oxygen. When the singlet oxygen diffuses around the donor beads and reaches the acceptor beads located in close proximity, a chemiluminescent reaction within the acceptor beads is induced. This reaction ultimately results in light emission. If molecules linked to the donor beads do not interact with molecules linked to the acceptor beads, the singlet oxygen produced by donor beads do not reach the acceptor beads and chemiluminescent reaction does not occur.


For example, a biotin-labeled antigen-binding molecule comprising Fc region is immobilized to the donor beads and glutathione S-transferase (GST)-tagged Fcγ receptor is immobilized to the acceptor beads. In the absence of an antigen-binding molecule comprising a competitive Fc region variant, Fcγ receptor interacts with a polypeptide complex comprising a wild-type Fc region, inducing a signal of 520 to 620 nm as a result. The antigen-binding molecule having a non-tagged Fc region variant competes with the antigen-binding molecule comprising a native Fc region for the interaction with Fcγreceptor. The relative binding affinity can be determined by quantifying the reduction of fluorescence as a result of competition. Methods for biotinylating the antigen-binding molecules such as antibodies using Sulfo-NHS-biotin or the like are known. Appropriate methods for adding the GST tag to an Fcγreceptor include methods that involve fusing polypeptides encoding Fcγ and GST in-frame, expressing the fused gene using cells introduced with a vector to which the gene is operably linked, and then purifying using a glutathione column. The induced signal can be preferably analyzed, for example, by fitting to a one-site competition model based on nonlinear regression analysis using software such as GRAPHPAD PRISM (GraphPad; San Diego).


One of the substances for observing their interaction is immobilized as a ligand onto the gold thin layer of a sensor chip. When light is shed on the rear surface of the sensor chip so that total reflection occurs at the interface between the gold thin layer and glass, the intensity of reflected light is partially reduced at a certain site (SPR signal). The other substance for observing their interaction is injected as an analyte onto the surface of the sensor chip. The mass of immobilized ligand molecule increases when the analyte binds to the ligand. This alters the refraction index of solvent on the surface of the sensor chip. The change in refraction index causes a positional shift of SPR signal (conversely, the dissociation shifts the signal back to the original position). In the Biacore system, the amount of shift described above (i.e., the change of mass on the sensor chip surface) is plotted on the vertical axis, and thus the change of mass over time is shown as measured data (sensorgram). Kinetic parameters (association rate constant (ka) and dissociation rate constant (kd)) are determined from the curve of sensorgram, and affinity (1(D) is determined from the ratio between these two constants Inhibition assay is preferably used in the BIACORE methods. Examples of such inhibition assay are described in Proc. Natl. Acad. Sci. USA (2006) 103(11), 4005-4010.


In addition to the Fc region of human IgG1 (SEQ ID NO: 9), IgG2 (SEQ ID NO: 10), IgG3 (SEQ ID NO: 11), or IgG4 (SEQ ID NO: 12), an Fc region with modified FcγR binding, which has a higher Fcγ receptor-binding activity than an Fc region of a native human IgG may be appropriately used as an Fc region included in the present invention. Herein, “Fc region of a native human IgG” refers to an Fc region in which the sugar chain bonded to position 297 (EU numbering) of the Fc region of human IgG1, IgG2, IgG3, or IgG4 shown in SEQ ID NO: 9, 10, 11, or 12 is a fucose-containing sugar chain. Such Fc regions with modified FcγR binding may be produced by altering amino acids of the Fc region of a native human IgG. Whether the FcγR-binding activity of an Fc region with modified FcγR binding is higher than that of an Fc region of a native human IgG can be determined appropriately using methods described in the abovementioned section “Binding Activity”.


In the present invention, “alteration of amino acids” or “amino acid alteration” of an Fc region includes alteration into an amino acid sequence which is different from that of the starting Fc region. The starting Fc region may be any Fc region, as long as a variant modified from the starting Fc region can bind to human Fcγ receptor in a neutral pH range. Furthermore, an Fc region modified from a starting Fc region which had been already modified can also be used preferably as an Fc region of the present invention. The “starting Fc region” can refer to the polypeptide itself, a composition comprising the starting Fc region, or an amino acid sequence encoding the starting Fc region. Starting Fc regions can comprise known Fc regions produced via recombination described briefly in the section “Antibody”. The origin of starting Fc regions is not limited, and they may be obtained from human or any nonhuman organisms. Such organisms preferably include mice, rats, guinea pigs, hamsters, gerbils, cats, rabbits, dogs, goats, sheep, bovines, horses, camels and organisms selected from nonhuman primates. In another embodiment, starting Fc regions can also be obtained from cynomolgus monkeys, marmosets, rhesus monkeys, chimpanzees, or humans. Starting Fc regions can be obtained preferably from human IgG1; however, they are not limited to any particular IgG class. This means that an Fc region of human IgG1, IgG2, IgG3, or IgG4 can be used appropriately as a starting Fc region, and herein also means that an Fc region of an arbitrary IgG class or subclass derived from any organisms described above can be preferably used as a starting Fc region. Examples of naturally-occurring IgG variants or modified forms are described in published documents (Curr. Opin. Biotechnol. (2009) 20 (6): 685-91; Curr. Opin. Immunol. (2008) 20 (4), 460-470; Protein Eng. Des. Sel. (2010) 23 (4): 195-202; International Publication Nos. WO 2009/086320, WO 2008/092117, WO 2007/041635, and WO 2006/105338); however, they are not limited to the examples.


Examples of alterations include those with one or more mutations, for example, mutations by substitution of different amino acid residues for amino acids of starting Fc regions, by insertion of one or more amino acid residues into starting Fc regions, or by deletion of one or more amino acids from starting Fc region. Preferably, the amino acid sequences of altered Fc regions comprise at least a part of the amino acid sequence of a non-native Fc region. Such variants necessarily have sequence identity or similarity less than 100% to their starting Fc region. In a preferred embodiment, the variants have amino acid sequence identity or similarity about 75% to less than 100%, more preferably about 80% to less than 100%, even more preferably about 85% to less than 100%, still more preferably about 90% to less than 100%, and yet more preferably about 95% to less than 100% to the amino acid sequence of their starting Fc region. In a non-limiting embodiment of the present invention, at least one amino acid is different between an FcγR-binding modified Fc region of the present invention and its starting Fc region. Amino acid difference between an FcγR-binding modified Fc region of the present invention and its starting Fc region can also be preferably specified based on the specific amino acid differences at the above-described specific amino acid positions according to EU numbering system.


Known methods such as site-directed mutagenesis (Kunkel et al. (Proc. Natl. Acad. Sci. USA (1985) 82, 488-492)) and Overlap extension PCR can be appropriately employed to alter the amino acids of Fc regions. Furthermore, various known methods can also be used as an amino acid alteration method for substituting amino acids by those other than natural amino acids (Annu Rev. Biophys. Biomol. Struct. (2006) 35, 225-249; Proc. Natl. Acad. Sci. U.S.A. (2003) 100 (11), 6353-6357). For example, a cell-free translation system (Clover Direct (Protein Express)) containing tRNAs in which amber suppressor tRNA, which is complementary to UAG codon (amber codon) that is a stop codon, is linked with an unnatural amino acid may be suitably used.


Included in the antigen-binding molecules of the present invention, an Fc region with modified FcγR binding, which has a higher Fcγ receptor-binding activity than that of an Fc region of a native human IgG, (an FcγR binding-modified Fc region) may be obtained by any method. Specifically, the Fc region with modified FcγR binding may be obtained by altering amino acids of an IgG-type human immunoglobulin used as a starting Fc region. Preferred Fc regions of the IgG-type immunoglobulins for alteration include, for example, those of human IgGs shown in SEQ ID NO: 9, 10, 11, or 12 (IgG1, IgG2, IgG3, or IgG4, respectively, and variants thereof).


Amino acids of any positions may be altered to other amino acids, as long as the binding activity toward the Fcγ receptor is higher than that of the Fc region of a native human IgG. When the antigen-binding molecule contains a human IgG1 Fc region as the human Fc region, it preferably contains an alteration that yields the effect of a higher Fcγ receptor-binding activity than that of the Fc region of a native human IgG, in which the sugar chain bound at position 297 (EU numbering) is a fucose-containing sugar chain. Such amino acid alterations have been reported, for example, in international publications such as WO2007/024249, WO2007/021841, WO2006/031370, WO2000/042072, WO2004/029207, WO2004/099249, WO2006/105338, WO2007/041635, WO2008/092117, WO2005/070963, WO2006/020114, WO2006/116260, and WO2006/023403.


Examples of such amino acids that may be altered include at least one or more amino acids selected from the group consisting of positions 221, 222, 223, 224, 225, 227, 228, 230, 231, 232, 233, 234, 235, 236, 237, 238, 239, 240, 241, 243, 244, 245, 246, 247, 249, 250, 251, 254, 255, 256, 258, 260, 262, 263, 264, 265, 266, 267, 268, 269, 270, 271, 272, 273, 274, 275, 276, 278, 279, 280, 281, 282, 283, 284, 285, 286, 288, 290, 291, 292, 293, 294, 295, 296, 297, 298, 299, 300, 301, 302, 303, 304, 305, 311, 313, 315, 317, 318, 320, 322, 323, 324, 325, 326, 327, 328, 329, 330, 331, 332, 333, 334, 335, 336, 337, 339, 376, 377, 378, 379, 380, 382, 385, 392, 396, 421, 427, 428, 429, 434, 436, and 440 (EU numbering). An Fc region (Fc region with modified FcγR binding) having a higher Fcγ receptor-binding activity than that of an Fc region of a native human IgG can be obtained by altering these amino acids.


Examples of particularly preferable alterations for use in the present invention include at least one or more amino acid alterations selected from the group consisting of:


Lys or Tyr for the amino acid at position 221;


Phe, Trp, Glu, or Tyr for the amino acid at position 222;


Phe, Trp, Glu, or Lys for the amino acid at position 223;


Phe, Trp, Glu, or Tyr for the amino acid at position 224;


Glu, Lys, or Trp for the amino acid at position 225;


Glu, Gly, Lys, or Tyr for the amino acid at position 227;


Glu, Gly, Lys, or Tyr for the amino acid at position 228;


Ala, Glu, Gly, or Tyr for the amino acid at position 230;


Glu, Gly, Lys, Pro, or Tyr for the amino acid at position 231;


Glu, Gly, Lys, or Tyr for the amino acid at position 232;


Ala, Asp, Phe, Gly, His, Ile, Lys, Leu, Met, Asn, Gln, Arg, Ser, Thr, Val, Trp, or Tyr for the amino acid at position 233;


Ala, Asp, Glu, Phe, Gly, His, Ile, Lys, Met, Asn, Pro, Gln, Arg, Ser, Thr, Val, Trp, or Tyr for the amino acid at position 234;


Ala, Asp, Glu, Phe, Gly, His, Ile, Lys, Met, Asn, Pro, Gln, Arg, Ser, Thr, Val, Trp, or Tyr for the amino acid at position 235;


Ala, Asp, Glu, Phe, His, Ile, Lys, Leu, Met, Asn, Pro, Gln, Arg, Ser, Thr, Val, Trp, or Tyr for the amino acid at position 236;


Asp, Glu, Phe, His, Ile, Lys, Leu, Met, Asn, Pro, Gln, Arg, Ser, Thr, Val, Trp, or Tyr for the amino acid at position 237;


Asp, Glu, Phe, Gly, His, Ile, Lys, Leu, Met, Asn, Gln, Arg, Ser, Thr, Val, Trp, or Tyr for the amino acid at position 238;


Asp, Glu, Phe, Gly, His, Ile, Lys, Leu, Met, Asn, Pro, Gln, Arg, Thr, Val, Trp, or Tyr for the amino acid at position 239;


Ala, Ile, Met, or Thr for the amino acid at position 240;


Asp, Glu, Leu, Arg, Trp, or Tyr for the amino acid at position 241;


Leu, Glu, Leu, Gln, Arg, Trp, or Tyr for the amino acid at position 243;


His for the amino acid at position 244;


Ala for the amino acid at position 245;


Asp, Glu, His, or Tyr for the amino acid at position 246;


Ala, Phe, Gly, His, Ile, Leu, Met, Thr, Val, or Tyr for the amino acid at position 247;


Glu, His, Gln, or Tyr for the amino acid at position 249;


Glu or Gln for the amino acid at position 250;


Phe for the amino acid at position 251;


Phe, Met, or Tyr for the amino acid at position 254;


Glu, Leu, or Tyr for the amino acid at position 255;


Ala, Met, or Pro for the amino acid at position 256;


Asp, Glu, His, Ser, or Tyr for the amino acid at position 258;


Asp, Glu, His, or Tyr for the amino acid at position 260;


Ala, Glu, Phe, Ile, or Thr for the amino acid at position 262;


Ala, Ile, Met, or Thr for the amino acid at position 263;


Asp, Glu, Phe, Gly, His, Ile, Lys, Leu, Met, Asn, Pro, Gln, Arg, Ser, Thr, Trp, or Tyr for the amino acid at position 264;


Ala, Leu, Phe, Gly, His, Ile, Lys, Leu, Met, Asn, Pro, Gln, Arg, Ser, Thr, Val, Trp, or Tyr for the amino acid at position 265;


Ala, Ile, Met, or Thr for the amino acid at position 266;


Asp, Glu, Phe, His, Ile, Lys, Leu, Met, Asn, Pro, Gln, Arg, Thr, Val, Trp, or Tyr for the amino acid at position 267;


Asp, Glu, Phe, Gly, Ile, Lys, Leu, Met, Pro, Gln, Arg, Thr, Val, or Trp for the amino acid at position 268;


Phe, Gly, His, Ile, Lys, Leu, Met, Asn, Pro, Arg, Ser, Thr, Val, Trp, or Tyr for the amino acid at position 269;


Glu, Phe, Gly, His, Ile, Leu, Met, Pro, Gln, Arg, Ser, Thr, Trp, or Tyr for the amino acid at position 270;


Ala, Asp, Glu, Phe, Gly, His, Ile, Lys, Leu, Met, Asn, Gln, Arg, Ser, Thr, Val, Trp, or Tyr for the amino acid at position 271;


Asp, Phe, Gly, His, Ile, Lys, Leu, Met, Pro, Arg, Ser, Thr, Val, Trp, or Tyr for the amino acid at position 272;


Phe or Ile for the amino acid at position 273;


Asp, Glu, Phe, Gly, His, Ile, Leu, Met, Asn, Pro, Arg, Ser, Thr, Val, Trp, or Tyr for the amino acid at position 274;


Leu or Trp for the amino acid at position 275;


Asp, Glu, Phe, Gly, His, Ile, Leu, Met, Pro, Arg, Ser, Thr, Val, Trp, or Tyr for the amino acid at position 276;


Asp, Glu, Gly, His, Ile, Lys, Leu, Met, Asn, Pro, Gln, Arg, Ser, Thr, Val, or Trp for the amino acid at position 278;


Ala for the amino acid at position 279;


Ala, Gly, His, Lys, Leu, Pro, Gln, Trp, or Tyr for the amino acid at position 280;


Asp, Lys, Pro, or Tyr for the amino acid at position 281;


Glu, Gly, Lys, Pro, or Tyr for the amino acid at position 282;


Ala, Gly, His, Ile, Lys, Leu, Met, Pro, Arg, or Tyr for the amino acid at position 283;


Asp, Glu, Leu, Asn, Thr, or Tyr for the amino acid at position 284;


Asp, Glu, Lys, Gln, Trp, or Tyr for the amino acid at position 285;


Glu, Gly, Pro, or Tyr for the amino acid at position 286;


Asn, Asp, Glu, or Tyr for the amino acid at position 288;


Asp, Gly, His, Leu, Asn, Ser, Thr, Trp, or Tyr for the amino acid at position 290;


Asp, Glu, Gly, His, Ile, Gln, or Thr for the amino acid at position 291;


Ala, Asp, Glu, Pro, Thr, or Tyr for the amino acid at position 292;


Phe, Gly, His, Ile, Leu, Met, Asn, Pro, Arg, Ser, Thr, Val, Trp, or Tyr for the amino acid at position 293;


Phe, Gly, His, Ile, Lys, Leu, Met, Asn, Pro, Arg, Ser, Thr, Val, Trp, or Tyr for the amino acid at position 294;


Asp, Glu, Phe, Gly, His, Ile, Lys, Met, Asn, Pro, Arg, Ser, Thr, Val, Trp, or Tyr for the amino acid at position 295;


Ala, Asp, Glu, Gly, His, Ile, Lys, Leu, Met, Asn, Gln, Arg, Ser, Thr, or Val for the amino acid at position 296;


Asp, Glu, Phe, Gly, His, Ile, Lys, Leu, Met, Pro, Gln, Arg, Ser, Thr, Val, Trp, or Tyr for the amino acid at position 297;


Ala, Asp, Glu, Phe, His, Ile, Lys, Met, Asn, Gln, Arg, Thr, Val, Trp, or Tyr for the amino acid at position 298;


Ala, Asp, Glu, Phe, Gly, His, Ile, Lys, Leu, Met, Asn, Pro, Gln, Arg, Ser, Val, Trp, or Tyr for the amino acid at position 299;


Ala, Asp, Glu, Gly, His, Ile, Lys, Leu, Met, Asn, Pro, Gln, Arg, Ser, Thr, Val, or Trp for the amino acid at position 300;


Asp, Glu, His, or Tyr for the amino acid at position 301;


Ile for the amino acid at position 302;


Asp, Gly, or Tyr for the amino acid at position 303;


Asp, His, Leu, Asn, or Thr for the amino acid at position 304;


Glu, Ile, Thr, or Tyr for the amino acid at position 305;


Ala, Asp, Asn, Thr, Val, or Tyr for the amino acid at position 311;


Phe for the amino acid at position 313;


Leu for the amino acid at position 315;


Glu or Gln for the amino acid at position 317;


His, Leu, Asn, Pro, Gln, Arg, Thr, Val, or Tyr for the amino acid at position 318;


Asp, Phe, Gly, His, Ile, Leu, Asn, Pro, Ser, Thr, Val, Trp, or Tyr for the amino acid at position 320;


Ala, Asp, Phe, Gly, His, Ile, Pro, Ser, Thr, Val, Trp, or Tyr for the amino acid at position 322;


Ile for the amino acid at position 323;


Asp, Phe, Gly, His, Ile, Leu, Met, Pro, Arg, Thr, Val, Trp, or Tyr for the amino acid at position 324;


Ala, Asp, Glu, Phe, Gly, His, Ile, Lys, Leu, Met, Pro, Gln, Arg, Ser, Thr, Val, Trp, or Tyr for the amino acid at position 325;


Ala, Asp, Glu, Gly, Ile, Leu, Met, Asn, Pro, Gln, Ser, Thr, Val, Trp, or Tyr for the amino acid at position 326;


Ala, Asp, Glu, Phe, Gly, His, Ile, Lys, Leu, Met, Asn, Pro, Arg, Thr, Val, Trp, or Tyr for the amino acid at position 327;


Ala, Asp, Glu, Phe, Gly, His, Ile, Lys, Met, Asn, Pro, Gln, Arg, Ser, Thr, Val, Trp, or Tyr for the amino acid at position 328;


Asp, Glu, Phe, Gly, His, Ile, Lys, Leu, Met, Asn, Gln, Arg, Ser, Thr, Val, Trp, or Tyr for the amino acid at position 329;


Cys, Glu, Phe, Gly, His, Ile, Lys, Leu, Met, Asn, Pro, Arg, Ser, Thr, Val, Trp, or Tyr for the amino acid at position 330;


Asp, Phe, His, Ile, Leu, Met, Gln, Arg, Thr, Val, Trp, or Tyr for the amino acid at position 331;


Ala, Asp, Glu, Phe, Gly, His, Lys, Leu, Met, Asn, Pro, Gln, Arg, Ser, Thr, Val, Trp, or Tyr for the amino acid at position 332;


Ala, Asp, Glu, Phe, Gly, His, Ile, Leu, Met, Pro, Ser, Thr, Val, or Tyr for the amino acid at position 333;


Ala, Glu, Phe, Ile, Leu, Pro, or Thr for the amino acid at position 334;


Asp, Phe, Gly, His, Ile, Leu, Met, Asn, Pro, Arg, Ser, Val, Trp, or Tyr for the amino acid at position 335;


Glu, Lys, or Tyr for the amino acid at position 336;


Glu, His, or Asn for the amino acid at position 337;


Asp, Phe, Gly, Ile, Lys, Met, Asn, Gln, Arg, Ser, or Thr for the amino acid at position 339;


Ala or Val for the amino acid at position 376;


Gly or Lys for the amino acid at position 377;


Asp for the amino acid at position 378;


Asn for the amino acid at position 379;


Ala, Asn, or Ser for the amino acid at position 380;


Ala or Ile for the amino acid at position 382;


Glu for the amino acid at position 385;


Thr for the amino acid at position 392;


Leu for the amino acid at position 396;


Lys for the amino acid at position 421;


Asn for the amino acid at position 427;


Phe or Leu for the amino acid at position 428;


Met for the amino acid at position 429;


Trp for the amino acid at position 434;


Ile for the amino acid at position 436; and


Gly, His, Ile, Leu, or Tyr for the amino acid at position 440;


as indicated by EU numbering. The number of amino acids to be altered is not particularly limited; and amino acid may be altered at only one site or amino acids may be altered at two or more sites. Examples of combinations for amino acid alterations at two or more sites include those described in Table 6 (Tables 6-1 to 6-3).










TABLE 6-1





Combination of amino acids
Combination of amino acids







K370E/P396L/D270E
S239Q/I332Q


Q419H/P396L/D270E
S267D/I332E


V240A/P396L/D270E
S267E/I332E


R255L/P396L/D270E
S267L/A327S


R255L/P396L/D270E
S267Q/A327S


R255L/P396L/D270E/R292G
S298A/I332E


R255L/P396L/D270E
S304T/I332E


R255L/P396L/D270E/Y300L
S324G/I332D


F243L/D270E/K392N/P396L
S324G/I332E


F243L/R255L/D270E/P396L
S324I/I332D


F243L/R292P/Y300L/V305I/P396L
S324I/I332E


F243L/R292P/Y300L/P396L
T260H/I332E


F243L/R292P/Y300L
T335D/I332E


F243L/R292P/P396L
V240I/V266I


F243L/R292P/V305I
V264I/I332E


F243L/R292P
D265F/N297E/I332E


S298A/E333A/K334A
D265Y/N297D/I332E


E380A/T307A
F243L/V262I/V264W


K326M/E333S
N297D/A330Y/I332E


K326A/E333A
N297D/T299E/I332E


S317A/K353A
N297D/T299F/I332E


A327D/I332E
N297D/T299H/I332E


A330L/I332E
N297D/T299I/I332E


A330Y/I332E
N297D/T299L/I332E


E258H/I332E
N297D/T299V/I332E


E272H/I332E
P230A/E233D/I332E


E272I/N276D
P244H/P245A/P247V


E272R/I332E
S239D/A330L/I332E


E283H/I332E
S239D/A330Y/I332E


E293R/I332E
S239D/H268E/A330Y


F241L/V262I
S239D/I332E/A327A


F241W/F243W
S239D/I332E/A330I









Table 6-2 is a continuation of Table 6-1.












TABLE 6-2









F243L/V264I
S239D/N297D/I332E



H268D/A330Y
S239D/S298A/I332E



H268E/A330Y
S239D/V264I/I332E



K246H/I332E
S239E/N297D/I332E



L234D/I332E
S239E/V264I/I332E



L234E/I332E
S239N/A330L/I332E



L234G/I332E
S239N/A330Y/I332E



L234I/I332E
S239N/S298A/I332E



L234I/L235D
S239Q/V264I/I332E



L234Y/I332E
V264E/N297D/I332E



L235D/I332E
V264I/A330L/I332E



L235E/I332E
V264I/A330Y/I332E



L235I/I332E
V264I/S298A/I332E



L235S/I332E
Y296D/N297D/I332E



L328A/I332D
Y296E/N297D/I332E



L328D/I332D
Y296H/N297D/I332E



L328D/I332E
Y296N/N297D/I332E



L328E/I332D
Y296Q/N297D/I332E



L328E/I332E
Y296T/N297D/I332E



L328F/I332D
D265Y/N297D/T299L/I332E



L328F/I332E
F241E/F243Q/V262T/V264E



L328H/I332E
F241E/F243R/V262E/V264R



L328I/I332D
F241E/F243Y/V262T/V264R



L328I/I332E
F241L/F243L/V262I/V264I



L328M/I332D
F241R/F243Q/V262T/V264R



L328M/I332E
F241S/F243H/V262T/V264T



L328N/I332D
F241W/F243W/V262A/V264A



L328N/I332E
F241Y/F243Y/V262T/V264T



L328Q/I332D
I332E/A330Y/H268E/A327A



L328Q/I332E
N297D/I332E/S239D/A330L



L328T/I332D
N297D/S298A/A330Y/I332E



L328T/I332E
S239D/A330Y/I332E/K326E



L328V/I332D
S239D/A330Y/I332E/K326T



L328V/I332E
S239D/A330Y/I332E/L234I



L328Y/I332D
S239D/A330Y/I332E/L235D










Table 6-3 is a continuation of Table 6-2.












TABLE 6-3









L328Y/I332E
S239D/A330Y/I332E/V240I



N297D/I332E
S239D/A330Y/I332E/V264T



N297E/I332E
S239D/A330Y/I332E/V266I



N297S/I332E
S239D/D265F/N297D/I332E



P227G/I332E
S239D/D265H/N297D/I332E



P230A/E233D
S239D/D265I/N297D/I332E



Q295E/I332E
S239D/D265L/N297D/I332E



R255Y/I332E
S239D/D265T/N297D/I332E



S239D/I332D
S239D/D265V/N297D/I332E



S239D/I332E
S239D/D265Y/N297D/I332E



S239D/I332N
S239D/I332E/A330Y/A327A



S239D/I332Q
S239D/I332E/H268E/A327A



S239E/D265G
S239D/I332E/H268E/A330Y



S239E/D265N
S239D/N297D/I332E/A330Y



S239E/D265Q
S239D/N297D/I332E/K326E



S239E/I332D
S239D/N297D/I332E/L235D



S239E/I332E
S239D/V264I/A330L/I332E



S239E/I332N
S239D/V264I/S298A/I332E



S239E/I332Q
S239E/V264I/A330Y/I332E



S239N/I332D
F241E/F243Q/V262T/V264E/I332E



S239N/I332E
F241E/F243R/V262E/V264R/I332E



S239N/I332N
F241E/F243Y/V262T/V264R/I332E



S239N/I332Q
F241R/F243Q/V262T/V264R/I332E



S239Q/I332D
S239D/I332E/H268E/A330Y/A327A



S239Q/I332E
S239E/V264I/S298A/A330Y/I332E



S239Q/I332N
F241Y/F243Y/V262T/V264T/N297D/I332E



S267E/L328F
G236D/S267E



S239D/S267E










For the pH conditions to measure the binding activity of the Fc region contained in the antigen-binding molecule of the present invention and the Fcγ receptor, conditions in an acidic pH range or in a neutral pH range may be suitably used. The neutral pH range, as a condition to measure the binding activity of the Fc region and the Fcγ receptor contained in the antigen-binding molecule of the present invention, generally indicates pH 6.7 to pH 10.0. Preferably, it is a range indicated with arbitrary pH values between pH 7.0 and pH8.0; and preferably, it is selected from pH 7.0, pH 7.1, pH 7.2, pH 7.3, pH 7.4, pH 7.5, pH 7.6, pH 7.7, pH 7.8, pH 7.9, and pH 8.0; and particularly preferably, it is pH 7.4, which is close to the pH of plasma (blood) in vivo. Herein, the acidic pH range, as a condition for having a binding activity of the Fc region and the Fcγ receptor contained in the antigen-binding molecule of the present invention, generally indicates pH 4.0 to pH 6.5. Preferably, it indicates pH 5.5 to pH 6.5, and particularly preferably, it indicates pH 5.8 to pH 6.0, which is close to the pH in the early endosome in vivo. With regard to the temperature used as a measurement condition, the binding affinity between an Fc region and an Fcγ receptor can be evaluated at any temperature between 10° C. and 50° C. Preferably, a temperature between 15° C. and 40° C. is used to determine the binding affinity between an Fc region and an Fcγ receptor. More preferably, any temperature between 20° C. and 35° C., such as any single temperature from 20° C., 21° C., 22° C., 23° C., 24° C., 25° C., 26° C., 27° C., 28° C., 29° C., 30° C., 31° C., 32° C., 33° C., 34° C., and 35° C., can be similarly used to determine the binding affinity between an Fc region and an Fcγ receptor. A temperature of 25° C. is a non-limiting example in an embodiment of the present invention.


Herein, “Fc region with modified FcγR binding has a higher Fcγ receptor-binding activity than the native Fc region” means that the human Fcγ receptor-binding activity of the Fc region with modified FcγR binding toward any of the human Fcγ receptors of FcγRI, FcγRIIa, FcγRIIb, FcγRIIIa, and/or FcγRIIIb is higher than the binding activity of the native Fc region toward these human Fcγ receptors. For example, it means that based on an above-described analytical method, in comparison to the binding activity of an antigen-binding molecule containing a native human IgG Fc region as a control, the binding activity of the antigen-binding molecule comprising an Fc region with modified FcγR binding is 105% or more, preferably 110% or more, 115% or more, 120% or more, 125% or more, particularly preferably 130% or more, 135% or more, 140% or more, 145% or more, 150% or more, 155% or more, 160% or more, 165% or more, 170% or more, 175% or more, 180% or more, 185% or more, 190% or more, 195% or more, 2-fold or more, 2.5-fold or more, 3-fold or more, 3.5-fold or more, 4-fold or more, 4.5-fold or more, 5-fold or more, 7.5-fold or more, 10-fold or more, 20-fold or more, 30-fold or more, 40-fold or more, 50-fold or more, 60-fold or more, 70-fold or more, 80-fold or more, 90-fold or more, or 100-fold or more. The starting Fc region may be used as a native Fc region, and native Fc regions of antibodies of the same subclass may also be used.


In the present invention, an Fc region of a native human IgG in which the sugar chain bonded to the amino acid at position 297 (EU numbering) is a fucose-containing sugar chain, is suitably used as a native Fc region of human IgG to be used as a control. Whether or not the sugar chain bonded to the amino acid at position 297 (EU numbering) is a fucose-containing sugar chain can be determined using the technique described in Non-patent Document 6. For example, it is possible to determine whether or not the sugar chain bonded to the native human IgG Fc region is a fucose-containing sugar chain by a method such as the one below. Sugar chain is dissociated from a native human IgG to be tested, by reacting the test native human IgG with N-Glycosidase F (Roche diagnostics) (Weitzhandler et al. (J. Pharma. Sciences (1994) 83, 12, 1670-1675)). Next, a dried concentrate of a reaction solution from which protein has been removed by reaction with ethanol (Schenk et al. (J. Clin. Investigation (2001) 108 (11) 1687-1695)) is fluorescently labeled with 2-aminopyridine (Bigge et al. (Anal. Biochem. (1995) 230 (2) 229-238)). Reagents are removed by solid extraction using a cellulose cartridge, and the fluorescently labeled 2-AB-modified sugar chain is analyzed by normal-phase chromatography. It is possible to determine whether or not the sugar chain bonded to the native Fc region of a human IgG is a fucose-containing sugar chain by observing the detected chromatogram peaks.


As an antigen-binding molecule containing an Fc region of a native antibody of the same subclass, which is to be used as a control, an antigen-binding molecule having an Fc region of a monoclonal IgG antibody may be suitably used. The structures of the Fc regions are described in SEQ ID NO: 9 (A is added to the N terminus of RefSeq Accession No. AAC82527.1), SEQ ID NO: 10 (A is added to the N terminus of RefSeq Accession No. AAB59393.1), SEQ ID NO: 11 (RefSeq Accession No. CAA27268.1), and SEQ ID NO: 12 (A is added to the N terminus of RefSeq Accession No. AAB59394.1). Further, when an antigen-binding molecule containing an Fc region of a particular antibody isotype is used as the test substance, the effect of the antigen-binding molecule containing the test Fc region on Fcγ receptor-binding activity is tested by using as a control an antigen-binding molecule having an Fc region of a monoclonal IgG antibody of that particular isotype. In this way, antigen-binding molecules containing an Fc region of which Fcγ receptor-binding activity is demonstrated to be high are suitably selected.


Fc Regions Having a Selective Binding Activity to an Fcγ Receptor

Examples of Fc regions suitable for use in the present invention include Fc regions having a higher binding activity to a particular Fcγ receptor than to other Fcγ receptors (Fc regions having a selective binding activity to an Fcγ receptor). When an antibody is used as the antigen-binding molecule, a single antibody molecule can only bind to a single Fcγ receptor molecule. Therefore, a single antigen-binding molecule cannot bind to other activating FcγRs in an inhibitory Fcγ receptor-bound state, and cannot bind to other activating Fcγ receptors or inhibitory Fcγ receptors in an activating Fcγ receptor-bound state.


As described above, suitable examples of activating Fcγ receptors include FcγRI (CD64) which includes FcγRIa, FcγRIb, and FcγRIc; FcγRIII (CD16) which includes isoforms FcγRIIIa (including allotypes V158 and F158) and FcγRIIIb (including allotypes FcγRIIIb-NA1 and FcγRIIIb-NA2); and FcγRIIa (including allotypes H131 and R131). Meanwhile, suitable examples of inhibitory Fcγ receptors include FcγRIIb (including FcγRIIb-1 and FcγRIIb-2).


Herein, an example of a case where the binding activity toward a certain Fcγ receptor is higher than the binding activity toward another Fcγ receptor is the case where the binding activity toward an inhibitory Fcγ receptor is higher than the binding activity toward an activating Fcγ receptor. In this case, the binding activity of the Fc region toward FcγRIIb is said to be higher than the binding activity toward any of the human Fcγ receptors of FcγRIa, FcγRIIa, FcγRIIIa, and/or FcγRIIIb. For example, this means that, based on an above-described analytical method, the binding activity of an antigen-binding molecule containing the Fc region toward FcγRIIb is 105% or more, preferably 110% or more, 120% or more, 130% or more, 140% or more, particularly preferably 150% or more, 160% or more, 170% or more, 180% or more, 190% or more, 200% or more, 250% or more, 300% or more, 350% or more, 400% or more, 450% or more, 500% or more, 750% or more, 10-fold or more, 20-fold or more, 30-fold or more, 40-fold or more, 50-fold, 60-fold, 70-fold, 80-fold, 90-fold, or 100-fold or more as compared with the binding activity toward any of the human Fcγ receptors of FcγRIa, FcγRIIa, FcγRIIIa, and/or FcγRIIIb.


In a non-limiting embodiment of the present invention, a preferred example of an Fc region having a higher inhibitory Fcγ receptor-binding activity than an activating Fcγ receptor-binding activity (having a selective binding activity toward an inhibitory Fcγ receptor) is an Fc region in which the amino acid at position 238 or 328 as indicated by EU numbering in the aforementioned Fc region has been altered to an amino acid different from that of the native Fc region.


In a non-limiting embodiment of the present invention, a suitable example of an Fc region that has a higher binding activity toward an inhibitory Fcγ receptor than toward an activating Fcγ receptor (i.e., having a selective binding activity toward an inhibitory Fcγ receptor) is an Fc region with one or more of the following alterations in the amino acids (indicated by EU numbering) of the aforementioned Fc region: the amino acid at position 238 is altered to Asp and the amino acid at position 328 is altered to Glu. The Fc regions and alterations described in US2009/0136485 may be selected appropriately as the Fc region having a selective binding activity to an inhibitory Fcγ receptor.


In a non-limiting embodiment of the present invention, a suitable example is an Fc region in which one or more of the amino acids indicated by EU numbering at positions 238 and 328 according to EU numbering are respectively altered to Asp or Glu in the aforementioned Fc region.


Furthermore, in a non-limiting embodiment of the present invention, suitable examples of the Fc regions are those with substitution of Asp for Pro at position 238 (EU numbering), and one or more alterations selected from among Trp for the amino acid at position 237, Phe for the amino acid at position 237, Val for the amino acid at position 267, Gln for the amino acid at position 267, Asn for the amino acid at position 268, Gly for the amino acid at position 271, Leu for the amino acid at position 326, Gln for the amino acid at position 326, Glu for the amino acid at position 326, Met for the amino acid at position 326, Asp for the amino acid at position 239, Ala for the amino acid at position 267, Trp for the amino acid at position 234, Tyr for the amino acid at position 234, Ala for the amino acid at position 237, Asp for the amino acid at position 237, Glu for the amino acid at position 237, Leu for the amino acid at position 237, Met for the amino acid at position 237, Tyr for the amino acid at position 237, Lys for the amino acid at position 330, Arg for the amino acid at position 330, Asp for the amino acid at position 233, Asp for the amino acid at position 268, Glu for the amino acid at position 268, Asp for the amino acid at position 326, Ser for the amino acid at position 326, Thr for the amino acid at position 326, Ile for the amino acid at position 323, Leu for the amino acid at position 323, Met for the amino acid at position 323, Asp for the amino acid at position 296, Ala for the amino acid at position 326, Asn for the amino acid at position 326, and Met for the amino acid at position 330, according to EU numbering.


Antigen-Binding Molecule

In the present invention, an “antigen-binding molecule” is used in the broadest sense to refer to a molecule containing an antigen-binding domain and an Fc region. Specifically, the antigen-binding molecules include various types of molecules as long as they exhibit the antigen-binding activity. Molecules in which an antigen-binding domain is linked to an Fc region include, for example, antibodies. Antibodies may include single monoclonal antibodies (including agonistic antibodies and antagonistic antibodies), human antibodies, humanized antibodies, chimeric antibodies, and such. Alternatively, when used as antibody fragments, they preferably include antigen-binding domains and antigen-binding fragments (for example, Fab, F(ab′)2, scFv, and Fv). Scaffold molecules where three dimensional structures, such as already-known stable α/β barrel protein structure, are used as a scaffold (base) and only some portions of the structures are made into libraries to construct antigen-binding domains are also included in antigen-binding molecules of the present invention.


An antigen-binding molecule of the present invention may contain at least some portions of an Fc region that mediates the binding to FcRn and Fcγ receptor. In a non-limiting embodiment, the antigen-binding molecule includes, for example, antibodies and Fc fusion proteins. A fusion protein refers to a chimeric polypeptide comprising a polypeptide having a first amino acid sequence that is linked to a polypeptide having a second amino acid sequence that would not naturally link in nature. For example, a fusion protein may comprise the amino acid sequence encoding at least a portion of an Fc region (for example, a portion of an Fc region responsible for the binding to FcRn or a portion of an Fc region responsible for the binding to Fcγ receptor) and a non-immunoglobulin polypeptide containing, for example, the amino acid sequence encoding the ligand-binding domain of a receptor or a receptor-binding domain of a ligand. The amino acid sequences may be present in separate proteins that are transported together to a fusion protein, or generally may be present in a single protein; however, they are included in a new rearrangement in a fusion polypeptide. Fusion proteins can be produced, for example, by chemical synthesis, or by genetic recombination techniques to express a polynucleotide encoding peptide regions in a desired arrangement.


Each of the domains of the antigen-binding domain, Fc region, and such of the present invention can be linked together via linkers or directly via polypeptide binding. The linkers comprise arbitrary peptide linkers that can be introduced by genetic engineering, synthetic linkers, and linkers disclosed in, for example, Protein Engineering (1996) 9(3), 299-305. However, peptide linkers are preferred in the present invention. The length of the peptide linkers is not particularly limited, and can be suitably selected by those skilled in the art according to the purpose. The length is preferably five amino acids or more (without particular limitation, the upper limit is generally 30 amino acids or less, preferably 20 amino acids or less), and particularly preferably 15 amino acids.


For example, such peptide linkers preferably include:











Ser







Gly•Ser







Gly•Gly•Ser







Ser•Gly•Gly











(SEQ ID NO: 25)









Gly•Gly•Gly•Ser











(SEQ ID NO: 26)









Ser•Gly•Gly•Gly











(SEQ ID NO: 27)









Gly•Gly•Gly•Gly•Ser











(SEQ ID NO: 28)









Ser•Gly•Gly•Gly•Gly











(SEQ ID NO: 29)









Gly•Gly•Gly•Gly•Gly•Ser











(SEQ ID NO: 30)









Ser•Gly•Gly•Gly•Gly•Gly











(SEQ ID NO: 31)









Gly•Gly•Gly•Gly•Gly•Gly•Ser











(SEQ ID NO: 32)









Ser•Gly•Gly•Gly•Gly•Gly•Gly











(SEQ ID NO: 27)









(Gly•Gly•Gly•Gly•Ser)n











(SEQ ID NO: 28)









(Ser•Gly•Gly•Gly•Gly)n







[where n is an integer of 1 or larger]. The length or sequences of peptide linkers can be selected accordingly by those skilled in the art depending on the purpose.


Synthetic linkers (chemical crosslinking agents) is routinely used to crosslink peptides, and for example:


N-hydroxy succinimide (NHS),


disuccinimidyl suberate (DSS),


bis(sulfosuccinimidyl) suberate (BS3),


dithiobis(succinimidyl propionate) (DSP),


dithiobis(sulfosuccinimidyl propionate) (DTSSP),


ethylene glycol bis(succinimidyl succinate) (EGS),


ethylene glycol bis(sulfosuccinimidyl succinate) (sulfo-EGS),


disuccinimidyl tartrate (DST), disulfosuccinimidyl tartrate (sulfo-DST),


bis[2-(succinimidoxycarbonyloxy)ethyl]sulfone (BSOCOES),


and bis[2-(sulfosuccinimidoxycarbonyloxy)ethyl]sulfone (sulfo-BSOCOES). These crosslinking agents are commercially available.


When multiple linkers for linking the respective domains are used, they may all be of the same type, or may be of different types.


In addition to the linkers exemplified above, linkers with peptide tags such as His tag, HA tag, myc tag, and FLAG tag may also be suitably used. Furthermore, hydrogen bonding, disulfide bonding, covalent bonding, ionic interaction, and properties of binding with each other as a result of combination thereof may be suitably used. For example, the affinity between CH1 and CL of antibody may be used, and Fc regions originating from the above-described bispecific antibodies may also be used for hetero Fc region association. Moreover, disulfide bonds formed between domains may also be suitably used.


In order to link the respective domains via peptide linkage, polynucleotides encoding the domains are linked in frame. Known methods for linking polynucleotides in frame include techniques such as ligation of restriction fragments, fusion PCR, and overlapping PCR. Such methods can be appropriately used alone or in combination to produce the antigen-binding molecules of the present invention. In the present invention, the terms “linked” and “fused”, or “linkage” and “fusion” are used interchangeably. These terms mean that two or more elements or components such as polypeptides are linked together to form a single structure by any means including the above-described chemical linking means and recombination techniques. When two or more elements or components are polypeptides, fusing in frame means linking two or more units of reading frames to form a longer continuous reading frame while maintaining the correct reading frames of the polypeptides. When two molecules of Fab are used as the antigen-binding domain, an antibody which is an antigen-binding molecule of the present invention in which the antigen-binding domain is linked in frame to an Fc region by a peptide bond and not via a linker can be used as a suitable antigen-binding molecule of the present application.


Antigen-Binding Molecule which Eliminates Aggregated Antigens in Preference to Unaggregated Antigens from Plasma


By selecting an antigen-binding molecule with higher binding activity to an aggregated antigen than to an unaggregated antigen as the antigen-binding molecule of the present invention, its elimination effect can further be increased. Such antigen-binding molecules can be obtained from among antigen-binding molecules produced according to the later-described method of producing antigen-binding molecules by selecting antigen-binding molecules with larger difference in their binding activities for aggregated antigens and unaggregated antigens.


The elimination effect can also be enhanced by selecting antigen-binding molecules that bind to epitopes which are specific to aggregated antigens. Antigen-binding molecules that bind to such epitopes can be obtained using known methods. For example, they can be obtained by obtaining molecules that bind to an aggregated antigen, and then comparing binding of those antigen-binding molecules to the aggregated antigen and unaggregated antigen, and selecting antigen-binding molecules that have higher binding to the aggregated antigen (WO 2006/016644; EMBO J. (1994) 13, 1166-75; WO 2009/008529; and such).


Furthermore, the elimination effect can be enhanced by selecting an antigen-binding molecule that has a higher binding activity to an Fcγ receptor or to the FcRn of a complex formed between an aggregated antigen and an antigen-binding molecule than its binding activity to an Fcγ receptor or to the FcRn of a complex formed between an unaggregated antigen and the antigen-binding molecule. Such an antigen-binding molecule can be obtained, for example, by introducing into an antigen-binding molecule obtained according to the later-described method for producing antigen-binding molecules, alterations that yield the aforementioned effect of enhancing binding to an Fcγ receptor or an FcRn, allowing formation of complexes between the altered antigen-binding molecule and an aggregated antigen or between the altered antigen-binding molecule and an unaggregated antigen, and then selecting an antigen-binding molecule that shows large difference in its binding activity to the Fcγ receptor or FcRn of the complexes.


In the present invention, whether an aggregated antigen is eliminated in preference to an unaggregated antigen can be confirmed by comparing plasma clearance of the aggregated antigen and plasma clearance of the unaggregated antigen. Specifically, if the ratio of clearance of an aggregated antigen in the presence of a test antigen-binding molecule to clearance of an aggregated antigen in the absence of the antigen-binding molecule (clearance of an aggregated antigen in the presence of the antigen-binding molecule/clearance of an aggregated antigen in the absence of the antigen-binding molecule) is higher than the clearance ratio for the unaggregated antigen (clearance of unaggregated antigen in the presence of the antigen-binding molecule/clearance of unaggregated antigen in the absence of the antigen-binding molecule), the antigen-binding molecule can be determined to be eliminating the aggregated antigen in preference to the unaggregated antigen. In the present invention, the clearance ratio of the aggregated antigen is preferably at least 1.5 times the clearance ratio of the unaggregated antigen (clearance ratio for aggregated antigen/clearance ratio for unaggregated antigen).


In the present invention, as long as use of the antigen-binding molecule can eliminate the aggregated antigen from plasma, the embodiment of its use is not particularly limited. Examples of a non-limiting embodiment of such use are pharmaceutical compositions containing an antigen-binding molecule provided by the present invention and methods comprising administering to a subject an antigen-binding molecule provided by the present invention. An example of another non-limiting embodiment is use of the antigen-binding molecule in an ex vivo method for eliminating aggregated antigens from plasma, which includes contacting an immune complex formed through contact of an antigen-binding molecule of the present invention with plasma isolated from a subject, and containing the antigen-binding molecules and aggregated antigens with FcRn- or Fcγ receptor-expressing cells.


Improvement of Preference

In the present invention, “eliminate the aggregated antigen from plasma” refers to the ability of eliminating from plasma antigens present in the plasma when an antigen-binding molecule is administered in vivo or when the antigen-binding molecule is secreted in vivo. Therefore, in the present invention, one can say that “plasma clearance of aggregated antigen by the antigen-binding molecule takes place preferentially” when plasma clearance of aggregated antigen is promoted as compared to that of unaggregated antigen when the antigen-binding molecule is administered. Whether plasma clearance of aggregated antigen by the antigen-binding molecule is taking place preferentially can be determined, for example, by measuring the above-mentioned clearance ratio. The above-mentioned clearance ratio is measured for an antigen-binding molecule that can bind to an aggregated antigen and which shows change in antigen-binding activity according to ion concentration, such as decreased antigen-binding activity in an acidic pH range compared to antigen-binding activity in a neutral pH range (or decreased antigen-binding activity in a low calcium ion concentration compared to antigen-binding activity in a high calcium ion concentration); and when the difference relative to the clearance ratio for unaggregated antigen (clearance ratio (aggregated antigen)/clearance ratio (unaggregated antigen)) becomes large, one can determine that preference has been improved.


Furthermore, if the antigen-binding molecule is administered to or secreted in an organism having biological fluids in which aggregated and unaggregated antigens coexist, and this results in a larger decrease in plasma concentration of the aggregated antigen than the decrease in plasma concentration of the unaggregated antigen, in comparison to before administration of the antigen-binding molecule, one can determine that the preference has been improved.


Improvement of Pharmacokinetics

In the present invention, “enhancement of pharmacokinetics”, “improvement of pharmacokinetics”, and “superior pharmacokinetics” can be restated as “enhancement of plasma (blood) retention”, “improvement of plasma (blood) retention”, “superior plasma (blood) retention”, and “prolonged plasma (blood) retention”. These terms are synonymous.


In the present invention, “improvement of pharmacokinetics” means not only prolongation of the period until elimination from the plasma (for example, until the antigen-binding molecule is degraded intracellularly or the like and cannot return to the plasma) after administration of the antigen-binding molecule to humans, or non-human animals such as mice, rats, monkeys, rabbits, and dogs, but also prolongation of the plasma retention of the antigen-binding molecule in a form that allows antigen binding (for example, in an antigen-free form of the antigen-binding molecule) during the period of administration to elimination due to degradation. Human IgG having wild-type Fc region can bind to FcRn from non-human animals. For example, mouse can be preferably used to be administered in order to confirm the property of the antigen-binding molecule of the invention since human IgG having wild-type Fc region can bind to mouse FcRn stronger than to human FcRn (Int Immunol. (2001) 13(12): 1551-1559). As another example, mouse in which its native FcRn genes are disrupted and a transgene for human FcRn gene is harbored to be expressed (Methods Mol Biol. 2010; 602: 93-104) can also be preferably used to be administered in order to confirm the property of the antigen-binding molecule of the invention described hereinafter. Specifically, “improvement of pharmacokinetics” also includes prolongation of the period until elimination due to degradation of the antigen-binding molecule not bound to antigens (the antigen-free form of antigen-binding molecule). The antigen-binding molecule in plasma cannot bind to a new antigen if the antigen-binding molecule has already bound to an antigen. Thus, the longer the period that the antigen-binding molecule is not bound to an antigen, the longer the period that it can bind to a new antigen (the higher the chance of binding to another antigen). This enables reduction of the time period that an antigen is free of the antigen-binding molecule in vivo and prolongation of the period that an antigen is bound to the antigen-binding molecule. The plasma concentration of the antigen-free form of antigen-binding molecule can be increased and the period that the antigen is bound to the antigen-binding molecule can be prolonged by accelerating the antigen elimination from the plasma by administration of the antigen-binding molecule. Specifically, herein “improvement of the pharmacokinetics of antigen-binding molecule” includes the improvement of a pharmacokinetic parameter of the antigen-free form of the antigen-binding molecule (any of prolongation of the half-life in plasma, prolongation of mean retention time in plasma, and impairment of plasma clearance), prolongation of the period that the antigen is bound to the antigen-binding molecule after administration of the antigen-binding molecule, and acceleration of antigen-binding molecule-mediated antigen elimination from the plasma. The improvement of pharmacokinetics of antigen-binding molecule can be assessed by determining any one of the parameters, half-life in plasma, mean plasma retention time, and plasma clearance for the antigen-binding molecule or the antigen-free form thereof (“Pharmacokinetics: Enshu-niyoru Rikai (Understanding through practice)” Nanzando). For example, the plasma concentration of the antigen-binding molecule or antigen-free form thereof is determined after administration of the antigen-binding molecule to mice, rats, monkeys, rabbits, dogs, or humans. Then, each parameter is determined. When the plasma half-life or mean plasma retention time is prolonged, the pharmacokinetics of the antigen-binding molecule can be judged to be improved. The parameters can be determined by methods known to those skilled in the art. The parameters can be appropriately assessed, for example, by noncompartmental analysis using the pharmacokinetics analysis software WinNonlin (Pharsight) according to the appended instruction manual. The plasma concentration of antigen-free antigen-binding molecule can be determined by methods known to those skilled in the art, for example, using the assay method described in Clin Pharmacol. 2008 April; 48(4): 406-417.


In the present invention, “improvement of pharmacokinetics” also includes prolongation of the period that an antigen is bound to an antigen-binding molecule after administration of the antigen-binding molecule. Whether the period that an antigen is bound to the antigen-binding molecule after administration of the antigen-binding molecule is prolonged can be assessed by determining the plasma concentration of free antigen. The prolongation can be judged based on the determined plasma concentration of free antigen or the time period required for an increase in the ratio of free antigen concentration to the total antigen concentration.


The plasma concentration of free antigen not bound to the antigen-binding molecule or the ratio of free antigen concentration to the total antigen concentration can be determined by methods known to those skilled in the art, for example, by the method used in Pharm Res. (2006) January; 23(1): 95-103. Alternatively, when an antigen exhibits a particular function in vivo, whether the antigen is bound to an antigen-binding molecule that neutralizes the antigen function (antagonistic molecule) can be assessed by testing whether the antigen function is neutralized. Whether the antigen function is neutralized can be assessed by assaying an in vivo marker that reflects the antigen function. Whether the antigen is bound to an antigen-binding molecule that activates the antigen function (agonistic molecule) can be assessed by assaying an in vivo marker that reflects the antigen function.


Determination of the plasma concentration of free antigen and ratio of the amount of free antigen in plasma to the amount of total antigen in plasma, in vivo marker assay, and such measurements are not particularly limited; however, the assays are preferably carried out after a certain period of time has passed after administration of the antigen-binding molecule. In the present invention, the period after administration of the antigen-binding molecule is not particularly limited; those skilled in the art can determine the appropriate period depending on the properties and the like of the administered antigen-binding molecule. Such periods include, for example, one day after administration of the antigen-binding molecule, three days after administration of the antigen-binding molecule, seven days after administration of the antigen-binding molecule, 14 days after administration of the antigen-binding molecule, and 28 days after administration of the antigen-binding molecule. In the present invention, the concept “plasma antigen concentration” comprises both “total antigen concentration in plasma” which is the sum of antigen-binding molecule bound antigen and non-bound antigen concentration or “free antigen concentration in plasma” which is antigen-binding molecule non-bound antigen concentration.


The total antigen concentration in the plasma can be lowered by administration, as antigen-binding molecule, of the antigen-binding molecule of the present invention by 2-fold, 5-fold, 10-fold, 20-fold, 50-fold, 100-fold, 200-fold, 500-fold, 1,000-fold, or even higher as compared to administration of an antigen-binding molecule containing an antigen-binding domain whose antigen-binding activity is ion concentration-independent or an antigen-binding molecule containing an Fc region with an impaired FcγR-binding activity, or compared to when the antigen-binding domain molecule of the present invention is not administered.


Molar antigen/antigen-binding molecule ratio can be calculated as shown below:


value A: Molar antigen concentration at each time point


value B: Molar antigen-binding molecule concentration at each time point


value C: Molar antigen concentration per molar antigen-binding molecule concentration (molar antigen/antigen-binding molecule ratio) at each time point C=A/B.


Smaller value C indicates higher efficiency of antigen elimination per antigen-binding molecule whereas higher value C indicates lower efficiency of antigen elimination per antigen-binding molecule.


Administering an antigen-binding molecule of the present invention can lower the molar antigen/antigen-binding molecule ratio by 2-fold, 5-fold, 10-fold, 20-fold, 50-fold, 100-fold, 200-fold, 500-fold, 1000-fold or more as compared to administration of antigen-binding molecules that cannot form the immune complexes disclosed in the present invention, antigen-binding molecules containing antigen-binding domains of which antigen-binding activity is independent of ion concentrations, or antigen-binding molecules containing Fc regions with compromised binding activity toward FcγR or FcRn.


In the present invention, as reference for comparison with the antigen-binding molecules of the present invention, antigen-binding molecules that cannot form the immune complexes disclosed in the present invention, antigen-binding molecules containing antigen-binding domains of which antigen-binding activity is independent of ion concentrations, or antigen-binding molecules containing Fc regions with compromised binding activity toward FcγR or FcRn.


When an FcRn-mediated pathway is used in the incorporation of antigen-binding molecules of the present invention from the plasma into cells, reduction in the total antigen concentration in plasma or the molar antigen/antibody ratio can also be assessed using human FcRn transgenic mouse line 32 or line 276 (Jackson Laboratories, Methods Mol Biol. (2010) 602: 93-104) by either antigen-antibody co-injection model or steady-state antigen infusion model when the antigen-binding molecule do not cross-react to the mouse counterpart antigen. When the antigen-binding molecule cross-react with the mouse counterpart, they can also be assessed by simply injecting the antigen-binding molecule to human FcRn transgenic mouse line 32 or line 276 (Jackson Laboratories). In the co-injection model, mixture of the antigen-binding molecule and antigen is administered to mice. In the steady-state antigen infusion model, infusion pump filled with an antigen solution is embedded into mice to achieve a constant plasma antigen concentration, and then the antigen-binding molecule is injected to the mice. Test antigen-binding molecules are administered at the same dosage. The total antigen concentration in plasma, free antigen concentration in plasma, and antigen-binding molecule concentration in plasma are measured at appropriate time points using method known to those skilled in the art.


When an FcγR-mediated pathway is used in the incorporation of antigen-binding molecules of the present invention from the plasma into cells, reduction in the total antigen concentration in plasma or the molar antigen/antibody ratio can be assessed by either the antigen-antibody co-injection model or the steady-state antigen infusion model using the conventionally used C57BL/6J mice (Charles River Japan) when the antigen-binding molecule does not cross-react with the mouse counterpart antigen. If the antigen-binding molecule cross-reacts with the mouse counterpart, assessment can be carried out simply by injecting the antigen-binding molecule into the conventionally used C57BL/6J mice (Charles River Japan).


In the co-injection model, a mixture of the antigen-binding molecule and antigen is administered to mice. In the steady-state antigen infusion model, an infusion pump filled with an antigen solution is embedded into mice to achieve a constant plasma antigen concentration, and then the antigen-binding molecule is injected into the mice. Test antigen-binding molecules are administered at the same dose. The total antigen concentration in plasma, free antigen concentration in plasma, and antigen-binding molecule concentration in plasma are measured at appropriate time points using methods known to those skilled in the art.


Total or free antigen concentration in plasma and molar antigen/antigen-binding molecule ratio can be measured at 2, 4, 7, 14, 28, 56, or 84 days after administration to evaluate the long-term effect of the present invention. In other words, a long term plasma antigen concentration is determined by measuring total or free antigen concentration in plasma and molar antigen/antigen-binding molecule ratio at 2, 4, 7, 14, 28, 56, or 84 days after administration of an antigen-binding molecule in order to evaluate the property of the antigen-binding molecule of the present invention. Whether the reduction of plasma antigen concentration or molar antigen/antigen-binding molecule ratio is achieved by antigen-binding molecule described in the present invention can be determined by the evaluation of the reduction at any one or more of the time points described above.


Total or free antigen concentration in plasma and molar antigen/antigen-binding molecule ratio can be measured at 15 min, 1, 2, 4, 8, 12, or 24 hours after administration to evaluate the short-term effect of the present invention. In other words, a short term plasma antigen concentration is determined by measuring total or free antigen concentration in plasma and molar antigen/antigen-binding molecule ratio at 15 min, 1, 2, 4, 8, 12, or 24 hours after administration of an antigen-binding molecule in order to evaluate the property of the antigen-binding molecule of the present invention.


The route of administration of an antigen-binding molecule of the present invention can be selected from intradermal, intravenous, intravitreal, subcutaneous, intraperitoneal, parenteral and intramuscular injection.


In the present invention, it is preferable that the pharmacokinetics of the antigen-binding molecule in human is improved. When the plasma retention in human is difficult to determine, it may be predicted based on the plasma retention in mice (for example, normal mice, human antigen-expressing transgenic mice, human FcRn-expressing transgenic mice) or monkeys (for example, cynomolgus monkeys).


In the present invention, “the improvement of the pharmacokinetics and prolonged plasma retention of an antigen-binding molecule” means improvement of any pharmacokinetic parameter (any of prolongation of the half-life in plasma, prolongation of mean retention time in plasma, reduction of plasma clearance, and bioavailability) after in vivo administration of the antigen-binding molecule, or an increase in the concentration of the antigen-binding molecule in the plasma in an appropriate time after administration. It may be determined by measuring any parameter such as half-life in plasma, mean retention time in plasma, plasma clearance, and bioavailability of the antigen-binding molecule (Pharmacokinetics: Enshu-niyoru Rikai (Understanding through practice), (Nanzando)). For example, when an antigen-binding molecule is administered to mice (normal mice and human FcRn transgenic mice), rats, monkeys, rabbits, dogs, humans, and so on, and the concentration of the antigen-binding molecule in the plasma is determined and each of the parameters is calculated, the pharmacokinetics of the antigen-binding molecule can be judged to be improved when the plasma half-life or mean retention time in the plasma is prolonged. These parameters can be determined by methods known to those skilled in the art. For example, the parameters can be appropriately assessed by non-compartmental analysis using pharmacokinetics analysis software WinNonlin (Pharsight) according to the attached instruction manual.


While it is not bound to a particular theory, a mechanism such as that described in the Discussion of the later-described Examples is presented as an example of a mechanism that may take place when an antigen-binding molecule of the present invention, which binds to an aggregated antigen and comprises an Fc region and an antigen-binding domain whose antigen-binding activity varies depending on ion concentration, eliminates the aggregated antigen from plasma.


If an antibody that contains a native IgG1-type constant region against an aggregated antigen and shows pH- or Ca-dependent binding can form a large immune complex and bind to FcgR, FcRn, complement receptors, and such with avidity, it is thought that aggregated antigen elimination can be preferentially and greatly accelerated. It is thought that when GA2-IgG1 which binds to aggregated human IgA is administered, such large immune complexes are formed. It is thought that GA2-IgG1 was able to greatly accelerate elimination of aggregated human IgA because the immune complex comprising GA2-IgG1 and human IgA, which is an aggregated antigen bound to an Fc receptor such as FcRn or FcγR with avidity, and was quickly incorporated into Fc receptor-expressing cells. The IgA that dissociates from the immune complex in the endosomes of cells that have taken up the immune complex is degraded in the lysosomes. At the same time, the IgA-dissociated antibody, which was bound to FcRn in the endosomes, is subsequently recycled to the plasma and can bind again to IgA in the plasma. Elimination of human IgA in the plasma is thought to be greatly accelerated in this manner. A method using an amino-acid-variant of the Fc region which binds to FcRn in the pH neutral range is described in WO2011/122011 as a method for accelerating elimination of antigens from the plasma. The present invention is useful as a method for accelerating the elimination from the plasma of aggregated antigens without using the above-mentioned variants, and can further accelerate the elimination of the aggregated antigens from the plasma through combination with the above-mentioned variants. Moreover, aggregated antigens may be eliminated, other than from the plasma, from the interstitial fluid, synovial fluid, peritoneal fluid, pleural fluid, and pericardial fluid, as long as the cells contacting interstitial fluid, synovial fluid, peritoneal fluid, pleural fluid, or pericardial fluid express FcγR or FcRn. A non-limiting embodiment of such cells includes immune cells and such present in the interstitial fluid, synovial fluid, peritoneal fluid, pleural fluid, and pericardial fluid.


Ex Vivo Method for Eliminating Aggregated Antigens from the Plasma


An example of a non-limiting embodiment of the use of an antigen-binding molecule in the method provided by the present invention for eliminating aggregated antigens from the plasma includes use of the antigen-binding molecule in a so-called ex vivo method for eliminating aggregated antigens from the plasma, which includes forming an immune complex containing the antigen-binding molecules and aggregated antigens by allowing the antigen-binding molecules of the present invention to contact plasma isolated from a subject, and allowing the immune complex to contact cells expressing FcRn and/or Fcγ receptors.


Furthermore, an example of a non-limiting embodiment of the use of an antigen-binding molecule in the method provided by the present invention for eliminating aggregated antigens from plasma includes use of the antigen-binding molecule in a so-called ex vivo method for eliminating aggregated antigens from the plasma, which includes contacting an immune complex containing the antigen-binding molecules and aggregated antigens present in the plasma isolated from a subject to whom the antigen-binding molecules of the present invention are administered with cells expressing FcRn and/or Fcγ receptors.


Whether an aggregated antigen is eliminated in preference to an unaggregated antigen from plasma can be confirmed, for example, by comparing and evaluating the aforementioned plasma clearance ratio for aggregated antigen (clearance of aggregated antigen in the presence of the antigen-binding molecule/clearance of aggregated antigen in the absence of the antigen-binding molecule) and the clearance ratio for unaggregated antigen (clearance of unaggregated antigen in the presence of the antigen-binding molecule/clearance of unaggregated antigen in the absence of the antigen-binding molecule).


Method of Producing Antigen-Binding Molecules Containing an Fc Region and an Antigen-Binding Domain Whose Antigen-Binding Activity is Ion Concentration-Dependent

In a non-limiting embodiment of the present invention, after isolating a polynucleotide encoding an antigen-binding domain whose binding activity changes depending on the condition selected as described above, the polynucleotide is inserted into an appropriate expression vector. For example, when the antigen-binding domain is an antibody variable region, once a cDNA encoding the variable region is obtained, the cDNA is digested with restriction enzymes that recognize the restriction sites inserted at the two ends of the cDNA. Preferably, the restriction enzymes recognize and digest a nucleotide sequence that appears at a low frequency in the nucleotide sequence composing the gene of the antigen-binding molecule. Furthermore, restriction enzymes that provide cohesive ends are preferably inserted to insert a single copy of a digested fragment into the vector in the correct orientation. The cDNA encoding a variable region of an antigen-binding molecule digested as described above is inserted into an appropriate expression vector to obtain an expression vector for the antigen-binding molecule of the present invention. At this time, a gene encoding an antibody constant region (C region) may be fused in frame with the gene encoding the variable region.


To produce an antigen-binding molecule of interest, a polynucleotide encoding the antigen-binding molecule is inserted in a manner operably linked to a regulatory sequence into an expression vector. Regulatory sequences include, for example, enhancers and promoters. Furthermore, an appropriate signal sequence may be linked to the amino terminus so that the expressed antigen-binding molecule is secreted to the outside of the cells. As signal sequence, for example, a peptide having the amino acid sequence MGWSCIILFLVATATGVHS (SEQ ID NO: 1) is used; however, it is also possible to link other appropriate signal sequences. The expressed polypeptide is cleaved at the carboxyl terminus of the above-described sequence, and the cleaved polypeptide is secreted as a mature polypeptide to the outside of cells. Then, appropriate host cells are transformed with this expression vector so that recombinant cells expressing the polynucleotide encoding the antigen-binding molecule of interest can be obtained. The antigen-binding molecules of the present invention can be produced from the recombinant cells by following the methods described above in the section “Antibody”.


For a nucleic acid, “operably linked” means that the nucleic acid has a functional relationship with another nucleic acid sequence. For example, a DNA encoding a presequence or a secretory leader is operably linked to a DNA encoding a certain polypeptide if it is to be expressed as a precursor protein involved in the secretion of the polypeptide. A promoter or enhancer is operably linked to a coding sequence if it affects the transcription of the coding sequence. A ribosome binding site is operably linked to a coding sequence if it is in a position that facilitates translation. Generally, “operably linked” means that the linked DNA sequences are contiguous, and in the case of a secretory leader, it means that the linked DNA sequences are contiguous and in a reading frame. However, enhancers do not have to be contiguous. Linking is accomplished by ligation at suitable restriction sites. If such sites do not exist, synthetic oligonucleotide adaptors or linkers are used in accordance with conventional practice. Furthermore, linked nucleic acids may be produced by the above-mentioned overlap extension PCR technique.


In a non-limiting embodiment of the present invention, after isolating a polynucleotide encoding the above-described antigen-binding molecule whose binding activity varies depending on a selected condition, a variant of the polynucleotide is inserted into an appropriate expression vector. Such variants preferably include those prepared via humanization based on the polynucleotide sequence encoding an antigen-binding molecule of the present invention obtained by screening as a randomized variable region library a synthetic library or an immune library constructed originating from nonhuman animals. The same methods as described above for producing above-described humanized antibodies can be used as a method for producing humanized antigen-binding molecule variants.


In another embodiment, such variants preferably include those obtained by introducing an alteration that increases the antigen affinity (affinity maturation) of an antigen-binding molecule of the present invention into an isolated polynucleotide sequence for the molecule obtained by screening using a synthetic library or a naive library as a randomized variable region library. Such variants can be obtained by various known procedures for affinity maturation, including CDR mutagenesis (Yang et al. (J. Mol. Biol. (1995) 254, 392-403)), chain shuffling (Marks et al. (Bio/Technology (1992) 10, 779-783)), use of E. coli mutant strains (Low et al. (J. Mol. Biol. (1996) 250, 359-368)), DNA shuffling (Patten et al. (Curr. Opin. Biotechnol. (1997) 8, 724-733)), phage display (Thompson et al. (J. Mol. Biol. (1996) 256, 77-88)), and sexual PCR (Clameri et al. (Nature (1998) 391, 288-291)).


As described above, antigen-binding molecules that are produced by the production methods of the present invention include antigen-binding molecules having an Fc region. Various variants can be used as Fc regions. In an embodiment, variants of the present invention preferably include polynucleotides encoding antigen-binding molecules having a heavy chain in which a polynucleotide encoding an Fc region variant as described above is linked in frame to a polynucleotide encoding the above-described antigen-binding molecule whose binding activity varies depending on a selected condition.


In a non-limiting embodiment of the present invention, Fc regions preferably include, for example, Fc constant regions of antibodies such as IgG1 of SEQ ID NO: 9 (Ala is added to the N terminus of AAC82527.1), IgG2 of SEQ ID NO: 10 (Ala is added to the N terminus of AAB59393.1), IgG3 of SEQ ID NO: 11 (CAA27268.1), and IgG4 of SEQ ID NO: 12 (Ala is added to the N terminus of AAB59394.1). A number of allotype sequences of human IgG1, human IgG2, human IgG3, and human IgG4 constant regions due to gene polymorphisms are described in “Sequences of proteins of immunological interest”, NIH Publication No. 91-3242. Any of such sequences may be used in the present invention. In particular, for the human IgG1 sequence, the amino acid sequence at positions 356 to 358 as indicated by EU numbering may be DEL or EEM. The plasma retention of IgG molecules is relatively long (the elimination from plasma is slow) since FcRn, particularly human FcRn, functions as a salvage receptor for IgG molecules. IgG molecules incorporated into endosomes by pinocytosis bind under the endosomal acidic condition to FcRn, particularly human FcRn, expressed in endosomes. IgG molecules that cannot bind to FcRn, particularly human FcRn, are transferred to lysosomes, and degraded there. Meanwhile, IgG molecules bound to FcRn, particularly human FcRn, are transferred to cell surface, and then return to plasma as a result of dissociation from FcRn, particularly human FcRn, under the neutral condition in plasma.


Since antibodies comprising a typical Fc region do not have a binding activity to FcRn, particularly to human FcRn, under the plasma neutral pH range condition, typical antibodies and antibody-antigen complexes are incorporated into cells by non-specific endocytosis and transferred to cell surface by binding to FcRn, particularly human FcRn, in the endosomal acidic pH range condition. FcRn, particularly human FcRn, transports antibodies from the endosome to the cell surface. Thus, some of FcRn, particularly human FcRn, is thought to be also present on the cell surface. However, antibodies are recycled to plasma, since they dissociated from FcRn, particularly human FcRn, in the neutral pH range condition on cell surface.


Fc regions having human FcRn-binding activity in the neutral pH range, which can be included in the antigen-binding molecules of the present invention, can be obtained by any method. Specifically, Fc regions having human FcRn-binding activity in the neutral pH range can be obtained by altering amino acids of human IgG-type immunoglobulin as a starting Fc region. Preferred Fc regions of human IgG-type immunoglobulin for alteration include, for example, those of human IgGs (IgG1, IgG2, IgG3, and IgG4, and variants thereof). Amino acids at any positions may be altered to other amino acids as long as the resulting regions have the human FcRn-binding activity in the neutral pH range or increased human FcRn-binding activity in the neutral range. When an antigen-binding molecule comprises the Fc region of human IgG1 as human Fc region, it is preferable that the resulting region comprises an alteration that results in the effect to enhance the human FcRn binding in the neutral pH range as compared to the binding activity of the starting Fc region of human IgG1. Amino acids that allow such alterations include, for example, amino acids at positions 221 to 225, 227, 228, 230, 232, 233 to 241, 243 to 252, 254 to 260, 262 to 272, 274, 276, 278 to 289, 291 to 312, 315 to 320, 324, 325, 327 to 339, 341, 343, 345, 360, 362, 370, 375 to 378, 380, 382, 385 to 387, 389, 396, 414, 416, 423, 424, 426 to 438, 440, and 442 (indicated by EU numbering). More specifically, such amino acid alterations include those listed in Table 5. Alteration of these amino acids enhances the human FcRn binding of the Fc region of IgG-type immunoglobulin in the neutral pH range.


Among those described above, appropriate alterations that enhance the human FcRn binding in the neutral pH range are selected for use in the present invention. Particularly preferred amino acids for such Fc region variants include, for example, amino acids at positions 237, 248, 250, 252, 254, 255, 256, 257, 258, 265, 286, 289, 297, 298, 303, 305, 307, 308, 309, 311, 312, 314, 315, 317, 332, 334, 360, 376, 380, 382, 384, 385, 386, 387, 389, 424, 428, 433, 434, and 436 (indicated by EU numbering). The human FcRn-binding activity of the Fc region included in an antigen-binding molecule can be increased in the neutral pH range by substituting at least one amino acid with a different amino acid.


Particularly preferred alterations in the Fc region include, for example, at least one or more amino acid alterations selected from the group of:


Met for the amino acid at position 237;


Ile for the amino acid at position 248;


Ala, Phe, Ile, Met, Gln, Ser, Val, Trp, or Tyr for the amino acid at position 250;


Phe, Trp, or Tyr for the amino acid at position 252;


Thr for the amino acid at position 254;


Glu for the amino acid at position 255;


Asp, Asn, Glu, or Gln for the amino acid at position 256;


Ala, Gly, Ile, Leu, Met, Asn, Ser, Thr, or Val for the amino acid at position 257;


His for the amino acid at position 258;


Ala for the amino acid at position 265;


Ala or Glu for the amino acid at position 286;


His for the amino acid at position 289;


Ala for the amino acid at position 297;


Ala for the amino acid at position 303;


Ala for the amino acid at position 305;


Ala, Asp, Phe, Gly, His, Ile, Lys, Leu, Met, Asn, Pro, Gln, Arg, Ser, Val, Trp, or Tyr for the amino acid at position 307;


Ala, Phe, Ile, Leu, Met, Pro, Gln, or Thr for the amino acid at position 308;


Ala, Asp, Glu, Pro, or Arg for the amino acid at position 309;


Ala, His, or Ile for the amino acid at position 311;


Ala or His for the amino acid at position 312;


Lys or Arg for the amino acid at position 314;


Ala, Asp, or His for the amino acid at position 315;


Ala for the amino acid at position 317;


Val for the amino acid at position 332;


Leu for the amino acid at position 334;


His for the amino acid at position 360;


Ala for the amino acid at position 376;


Ala for the amino acid at position 380;


Ala for the amino acid at position 382;


Ala for the amino acid at position 384;


Asp or His for the amino acid at position 385;


Pro for the amino acid at position 386;


Glu for the amino acid at position 387;


Ala or Ser for the amino acid at position 389;


Ala for the amino acid at position 424;


Ala, Asp, Phe, Gly, His, Ile, Lys, Leu, Asn, Pro, Gln, Ser, Thr, Val, Trp, or Tyr for the amino acid at position 428;


Lys for the amino acid at position 433;


Ala, Phe, His, Ser, Trp, or Tyr for the amino acid at position 434; and


His, Ile, Leu, Phe, Thr, or Val for the amino acid at position 436 in the EU numbering system. Meanwhile, the number of altered amino acids is not particularly limited; such amino acid alterations include single amino acid alteration and alteration of amino acids at two or more sites. Combinations of amino acid alterations at two or more sites include, for example, those described in Tables 5-1 to 5-32.


In addition to the Fc region of human IgG1 (SEQ ID NO: 9), IgG2 (SEQ ID NO: 10), IgG3 (SEQ ID NO: 11), or IgG4 (SEQ ID NO: 12), as Fc regions included in the present invention, Fc regions with modified FcγR binding, which have a higher Fcγ receptor-binding activity than the Fc region of a native human IgG in which the sugar chain bound at position 297 (EU numbering) is a fucose-containing sugar chain, may be suitably used. Such Fc regions with modified FcγR binding may be produced by altering amino acids in the Fc region of a native human IgG. Whether the FcγR-binding activity of an Fc region is higher than that of the Fc region of a native human IgG, in which the sugar chain bound at position 297 (EU numbering) is a fucose-containing sugar chain, can be appropriately determined using methods such as those described above.


In the present invention, “alteration of amino acids” or “amino acid alteration” of an Fc region includes alteration into an amino acid sequence which is different from that of the starting Fc region. The starting Fc region may be any Fc region, as long as a variant altered from the starting Fc region can bind to human Fcγ receptor in a neutral pH range. Furthermore, an Fc region altered from a starting Fc region which had been already altered can also be used preferably as an Fc region of the present invention. The “starting Fc region” can refer to the polypeptide itself, a composition comprising the starting Fc region, or an amino acid sequence encoding the starting Fc region. Starting Fc regions can comprise a known Fc region produced via recombination described briefly in the section “Antibody”. The origin of starting Fc regions is not limited, and they may be obtained from human or any nonhuman organisms. Such organisms preferably include mice, rats, guinea pigs, hamsters, gerbils, cats, rabbits, dogs, goats, sheep, bovines, horses, camels and organisms selected from nonhuman primates. In another embodiment, starting Fc regions can also be obtained from cynomolgus monkeys, marmosets, rhesus monkeys, chimpanzees, or humans. Starting Fc regions can be obtained preferably from human IgG1; however, they are not limited to any particular IgG class. This means that an Fc region of human IgG1, IgG2, IgG3, or IgG4 can be used appropriately as a starting Fc region, and herein also means that an Fc region of an arbitrary IgG class or subclass derived from any organisms described above can be preferably used as a starting Fc region. Examples of naturally-occurring IgG variants or modified forms are described in published documents (Curr. Opin. Biotechnol. (2009) 20(6): 685-91; Curr. Opin. Immunol. (2008) 20(4): 460-470; Protein Eng. Des. Sel. (2010) 23(4): 195-202; International Publication Nos. WO 2009/086320, WO 2008/092117, WO 2007/041635, and WO 2006/105338); however, they are not limited to the examples.


Examples of alterations include those with one or more mutations, for example, mutations by substitution of different amino acid residues for amino acids of starting Fc regions, by insertion of one or more amino acid residues into starting Fc regions, or by deletion of one or more amino acids from starting Fc region. Preferably, the amino acid sequences of altered Fc regions comprise at least a part of the amino acid sequence of a non-native Fc region. Such variants necessarily have sequence identity or similarity less than 100% to their starting Fc region. In a preferred embodiment, the variants have amino acid sequence identity or similarity about 75% to less than 100%, more preferably about 80% to less than 100%, even more preferably about 85% to less than 100%, still more preferably about 90% to less than 100%, and yet more preferably about 95% to less than 100% to the amino acid sequence of their starting Fc region. In a non-limiting embodiment of the present invention, at least one amino acid is different between an FcγR-binding modified Fc region of the present invention and its starting Fc region. The amino acid differences between an FcγR-binding modified Fc region of the present invention and its starting Fc region can also be suitably specified based on the amino acid differences at the above-described particular amino acid positions specified by the EU numbering system.


The Fc region with modified FcγR binding, which has a higher Fcγ receptor-binding activity than that of the Fc region of a native human IgG in which the sugar chain bound at position 297 (EU numbering) is a fucose-containing sugar chain, contained in the antigen-binding molecules of the present invention may be obtained by any method. Specifically, the Fc region with modified FcγR binding can be obtained by altering amino acids in a human IgG-type immunoglobulin, and is used as the starting Fc region. Preferred Fc regions of IgG-type immunoglobulins for alteration include, for example, Fc regions of human IgGs (IgG1, IgG2, IgG3, IgG4, and variants thereof).


Amino acids of any positions may be altered to other amino acids, as long as the binding activity toward the Fcγ receptor is higher than that of the Fc region of a native human IgG, in which the sugar chain bound at position 297 (EU numbering) is a fucose-containing sugar chain. When the antigen-binding molecule contains a human IgG1 Fc region as the human Fc region, it preferably contains an alteration that yields the effect of a higher Fcγ receptor-binding activity than that of the Fc region of a native human IgG, in which the sugar chain bound at position 297 (EU numbering) is a fucose-containing sugar chain. Such amino acid alterations have been reported, for example, in international publications such as WO 2007/024249, WO 2007/021841, WO 2006/031370, WO 2000/042072, WO 2004/029207, WO 2004/099249, WO 2006/105338, WO 2007/041635, WO 2008/092117, WO 2005/070963, WO 2006/020114, WO 2006/116260, and WO 2006/023403.


Examples of such amino acids that can be altered include at least one or more amino acids selected from the group of positions 221, 222, 223, 224, 225, 227, 228, 230, 231, 232, 233, 234, 235, 236, 237, 238, 239, 240, 241, 243, 244, 245, 246, 247, 249, 250, 251, 254, 255, 256, 258, 260, 262, 263, 264, 265, 266, 267, 268, 269, 270, 271, 272, 273, 274, 275, 276, 278, 279, 280, 281, 282, 283, 284, 285, 286, 288, 290, 291, 292, 293, 294, 295, 296, 297, 298, 299, 300, 301, 302, 303, 304, 305, 311, 313, 315, 317, 318, 320, 322, 323, 324, 325, 326, 327, 328, 329, 330, 331, 332, 333, 334, 335, 336, 337, 339, 376, 377, 378, 379, 380, 382, 385, 392, 396, 421, 427, 428, 429, 434, 436, and 440 (EU numbering). Alteration of these amino acids can yield Fc regions (Fc regions with modified FcγR binding) having a higher Fcγ receptor-binding activity than the Fcγ receptor-binding activity of an Fc region of a native human IgG, in which the sugar chain bound at position 297 (EU numbering) is a fucose-containing sugar chain.


Examples of particularly preferred alterations for use in the present invention include at least one or more amino acid alterations in the Fc region selected from the group of:


Lys or Tyr for the amino acid at position 221;


Phe, Trp, Glu, or Tyr for the amino acid at position 222;


Phe, Trp, Glu, or Lys for the amino acid at position 223;


Phe, Trp, Glu, or Tyr for the amino acid at position 224;


Glu, Lys, or Trp for the amino acid at position 225;


Glu, Gly, Lys, or Tyr for the amino acid at position 227;


Glu, Gly, Lys, or Tyr for the amino acid at position 228;


Ala, Glu, Gly, or Tyr for the amino acid at position 230;


Glu, Gly, Lys, Pro, or Tyr for the amino acid at position 231;


Glu, Gly, Lys, or Tyr for the amino acid at position 232;


Ala, Asp, Phe, Gly, His, Ile, Lys, Leu, Met, Asn, Gln, Arg, Ser, Thr, Val, Trp, or Tyr for the amino acid at position 233;


Ala, Asp, Glu, Phe, Gly, His, Ile, Lys, Met, Asn, Pro, Gln, Arg, Ser, Thr, Val, Trp, or Tyr for the amino acid at position 234;


Ala, Asp, Glu, Phe, Gly, His, Ile, Lys, Met, Asn, Pro, Gln, Arg, Ser, Thr, Val, Trp, or Tyr for the amino acid at position 235;


Ala, Asp, Glu, Phe, His, Ile, Lys, Leu, Met, Asn, Pro, Gln, Arg, Ser, Thr, Val, Trp, or Tyr for the amino acid at position 236;


Asp, Glu, Phe, His, Ile, Lys, Leu, Met, Asn, Pro, Gln, Arg, Ser, Thr, Val, Trp, or Tyr for the amino acid at position 237;


Asp, Glu, Phe, Gly, His, Ile, Lys, Leu, Met, Asn, Gln, Arg, Ser, Thr, Val, Trp, or Tyr for the amino acid at position 238;


Asp, Glu, Phe, Gly, His, Ile, Lys, Leu, Met, Asn, Pro, Gln, Arg, Thr, Val, Trp, or Tyr for the amino acid at position 239;


Ala, Ile, Met, or Thr for the amino acid at position 240;


Asp, Glu, Leu, Arg, Trp, or Tyr for the amino acid at position 241;


Leu, Glu, Leu, Gln, Arg, Trp, or Tyr for the amino acid at position 243;


His for the amino acid at position 244;


Ala for the amino acid at position 245;


Asp, Glu, His, or Tyr for the amino acid at position 246;


Ala, Phe, Gly, His, Ile, Leu, Met, Thr, Val, or Tyr for the amino acid at position 247;


Glu, His, Gln, or Tyr for the amino acid at position 249;


Glu or Gln for the amino acid at position 250;


Phe for the amino acid at position 251;


Phe, Met, or Tyr for the amino acid at position 254;


Glu, Leu, or Tyr for the amino acid at position 255;


Ala, Met, or Pro for the amino acid at position 256;


Asp, Glu, His, Ser, or Tyr for the amino acid at position 258;


Asp, Glu, His, or Tyr for the amino acid at position 260;


Ala, Glu, Phe, Ile, or Thr for the amino acid at position 262;


Ala, Ile, Met, or Thr for the amino acid at position 263;


Asp, Glu, Phe, Gly, His, Ile, Lys, Leu, Met, Asn, Pro, Gln, Arg, Ser, Thr, Trp, or Tyr for the amino acid at position 264;


Ala, Leu, Phe, Gly, His, Ile, Lys, Leu, Met, Asn, Pro, Gln, Arg, Ser, Thr, Val, Trp, or Tyr for the amino acid at position 265;


Ala, Ile, Met, or Thr for the amino acid at position 266;


Asp, Glu, Phe, His, Ile, Lys, Leu, Met, Asn, Pro, Gln, Arg, Thr, Val, Trp, or Tyr for the amino acid at position 267;


Asp, Glu, Phe, Gly, Ile, Lys, Leu, Met, Pro, Gln, Arg, Thr, Val, or Trp for the amino acid at position 268;


Phe, Gly, His, Ile, Lys, Leu, Met, Asn, Pro, Arg, Ser, Thr, Val, Trp, or Tyr for the amino acid at position 269;


Glu, Phe, Gly, His, Ile, Leu, Met, Pro, Gln, Arg, Ser, Thr, Trp, or Tyr for the amino acid at position 270;


Ala, Asp, Glu, Phe, Gly, His, Ile, Lys, Leu, Met, Asn, Gln, Arg, Ser, Thr, Val, Trp, or Tyr for the amino acid at position 271;


Asp, Phe, Gly, His, Ile, Lys, Leu, Met, Pro, Arg, Ser, Thr, Val, Trp, or Tyr for the amino acid at position 272;


Phe or Ile for the amino acid at position 273;


Asp, Glu, Phe, Gly, His, Ile, Leu, Met, Asn, Pro, Arg, Ser, Thr, Val, Trp, or Tyr for the amino acid at position 274;


Leu or Trp for the amino acid at position 275;


Asp, Glu, Phe, Gly, His, Ile, Leu, Met, Pro, Arg, Ser, Thr, Val, Trp, or Tyr for the amino acid at position 276;


Asp, Glu, Gly, His, Ile, Lys, Leu, Met, Asn, Pro, Gln, Arg, Ser, Thr, Val, or Trp for the amino acid at position 278;


Ala for the amino acid at position 279;


Ala, Gly, His, Lys, Leu, Pro, Gln, Trp, or Tyr for the amino acid at position 280;


Asp, Lys, Pro, or Tyr for the amino acid at position 281;


Glu, Gly, Lys, Pro, or Tyr for the amino acid at position 282;


Ala, Gly, His, Ile, Lys, Leu, Met, Pro, Arg, or Tyr for the amino acid at position 283;


Asp, Glu, Leu, Asn, Thr, or Tyr for the amino acid at position 284;


Asp, Glu, Lys, Gln, Trp, or Tyr for the amino acid at position 285;


Glu, Gly, Pro, or Tyr for the amino acid at position 286;


Asn, Asp, Glu, or Tyr for the amino acid at position 288;


Asp, Gly, His, Leu, Asn, Ser, Thr, Trp, or Tyr for the amino acid at position 290;


Asp, Glu, Gly, His, Ile, Gln, or Thr for the amino acid at position 291;


Ala, Asp, Glu, Pro, Thr, or Tyr for the amino acid at position 292;


Phe, Gly, His, Ile, Leu, Met, Asn, Pro, Arg, Ser, Thr, Val, Trp, or Tyr for the amino acid at position 293;


Phe, Gly, His, Ile, Lys, Leu, Met, Asn, Pro, Arg, Ser, Thr, Val, Trp, or Tyr for the amino acid at position 294;


Asp, Glu, Phe, Gly, His, Ile, Lys, Met, Asn, Pro, Arg, Ser, Thr, Val, Trp, or Tyr for the amino acid at position 295;


Ala, Asp, Glu, Gly, His, Ile, Lys, Leu, Met, Asn, Gln, Arg, Ser, Thr, or Val for the amino acid at position 296;


Asp, Glu, Phe, Gly, His, Ile, Lys, Leu, Met, Pro, Gln, Arg, Ser, Thr, Val, Trp, or Tyr for the amino acid at position 297;


Ala, Asp, Glu, Phe, His, Ile, Lys, Met, Asn, Gln, Arg, Thr, Val, Trp, or Tyr for the amino acid at position 298;


Ala, Asp, Glu, Phe, Gly, His, Ile, Lys, Leu, Met, Asn, Pro, Gln, Arg, Ser, Val, Trp, or Tyr for the amino acid at position 299;


Ala, Asp, Glu, Gly, His, Ile, Lys, Leu, Met, Asn, Pro, Gln, Arg, Ser, Thr, Val, or Trp for the amino acid at position 300;


Asp, Glu, His, or Tyr for the amino acid at position 301;


Ile for the amino acid at position 302;


Asp, Gly, or Tyr for the amino acid at position 303;


Asp, His, Leu, Asn, or Thr for the amino acid at position 304;


Glu, Ile, Thr, or Tyr for the amino acid at position 305;


Ala, Asp, Asn, Thr, Val, or Tyr for the amino acid at position 311;


Phe for the amino acid at position 313;


Leu for the amino acid at position 315;


Glu or Gln for the amino acid at position 317;


His, Leu, Asn, Pro, Gln, Arg, Thr, Val, or Tyr for the amino acid at position 318;


Asp, Phe, Gly, His, Ile, Leu, Asn, Pro, Ser, Thr, Val, Trp, or Tyr for the amino acid at position 320;


Ala, Asp, Phe, Gly, His, Ile, Pro, Ser, Thr, Val, Trp, or Tyr for the amino acid at position 322;


Ile for the amino acid at position 323;


Asp, Phe, Gly, His, Ile, Leu, Met, Pro, Arg, Thr, Val, Trp, or Tyr for the amino acid at position 324;


Ala, Asp, Glu, Phe, Gly, His, Ile, Lys, Leu, Met, Pro, Gln, Arg, Ser, Thr, Val, Trp, or Tyr for the amino acid at position 325;


Ala, Asp, Glu, Gly, Ile, Leu, Met, Asn, Pro, Gln, Ser, Thr, Val, Trp, or Tyr for the amino acid at position 326;


Ala, Asp, Glu, Phe, Gly, His, Ile, Lys, Leu, Met, Asn, Pro, Arg, Thr, Val, Trp, or Tyr for the amino acid at position 327;


Ala, Asp, Glu, Phe, Gly, His, Ile, Lys, Met, Asn, Pro, Gln, Arg, Ser, Thr, Val, Trp, or Tyr for the amino acid at position 328;


Asp, Glu, Phe, Gly, His, Ile, Lys, Leu, Met, Asn, Gln, Arg, Ser, Thr, Val, Trp, or Tyr for the amino acid at position 329;


Cys, Glu, Phe, Gly, His, Ile, Lys, Leu, Met, Asn, Pro, Arg, Ser, Thr, Val, Trp, or Tyr for the amino acid at position 330;


Asp, Phe, His, Ile, Leu, Met, Gln, Arg, Thr, Val, Trp, or Tyr for the amino acid at position 331;


Ala, Asp, Glu, Phe, Gly, His, Lys, Leu, Met, Asn, Pro, Gln, Arg, Ser, Thr, Val, Trp, or Tyr for the amino acid at position 332;


Ala, Asp, Glu, Phe, Gly, His, Ile, Leu, Met, Pro, Ser, Thr, Val, or Tyr for the amino acid at position 333;


Ala, Glu, Phe, Ile, Leu, Pro, or Thr for the amino acid at position 334;


Asp, Phe, Gly, His, Ile, Leu, Met, Asn, Pro, Arg, Ser, Val, Trp, or Tyr for the amino acid at position 335;


Glu, Lys, or Tyr for the amino acid at position 336;


Glu, His, or Asn for the amino acid at position 337;


Asp, Phe, Gly, Ile, Lys, Met, Asn, Gln, Arg, Ser, or Thr for the amino acid at position 339;


Ala or Val for the amino acid at position 376;


Gly or Lys for the amino acid at position 377;


Asp for the amino acid at position 378;


Asn for the amino acid at position 379;


Ala, Asn, or Ser for the amino acid at position 380;


Ala or Ile for the amino acid at position 382;


Glu for the amino acid at position 385;


Thr for the amino acid at position 392;


Leu for the amino acid at position 396;


Lys for the amino acid at position 421;


Asn for the amino acid at position 427;


Phe or Leu for the amino acid at position 428;


Met for the amino acid at position 429;


Trp for the amino acid at position 434;


Ile for the amino acid at position 436; and


Gly, His, Ile, Leu, or Tyr for the amino acid at position 440;


as indicated by EU numbering. Meanwhile, the number of amino acids to be altered is not particularly limited, and an amino acid at only one site may be altered or amino acids at two or more sites may be altered. Examples of combinations for the amino acid alterations at two or more sites include those described in Table 6 (Tables 6-1 to 6-3).


Among the Fc regions suitable for use in the present invention, a suitable example of an Fc region that has a higher binding activity toward an inhibitory Fcγ receptor than toward an activating Fcγ receptor (i.e., having a selective binding activity toward an inhibitory Fcγ receptor), which is used as a non-limiting embodiment of an Fc region with the property of having a higher binding activity toward a specific Fcγ receptor than toward other Fcγ receptors (i.e., an Fc region having a selective Fcγ receptor-binding activity), is an Fc region with one or more of the following alterations in the amino acids (indicated by EU numbering) of the aforementioned Fc region: the amino acid at position 238 is altered to Asp and the amino acid at position 328 is modified to Glu. The Fc regions and alterations described in US2009/0136485 may be selected appropriately as the Fc region having a selective binding activity to an inhibitory Fcγ receptor.


In a non-limiting embodiment of the present invention, a suitable example is an Fc region in which one or more of the amino acids indicated by EU numbering at positions 238 and 328 according to EU numbering are respectively altered to Asp or Glu in the aforementioned Fc region.


Furthermore, in a non-limiting embodiment of the present invention, suitable examples of the Fc regions are those with substitution of Asp for Pro at position 238 (EU numbering), and one or more alterations selected from among Trp for the amino acid at position 237, Phe for the amino acid at position 237, Val for the amino acid at position 267, Gln for the amino acid at position 267, Asn for the amino acid at position 268, Gly for the amino acid at position 271, Leu for the amino acid at position 326, Gln for the amino acid at position 326, Glu for the amino acid at position 326, Met for the amino acid at position 326, Asp for the amino acid at position 239, Ala for the amino acid at position 267, Trp for the amino acid at position 234, Tyr for the amino acid at position 234, Ala for the amino acid at position 237, Asp for the amino acid at position 237, Glu for the amino acid at position 237, Leu for the amino acid at position 237, Met for the amino acid at position 237, Tyr for the amino acid at position 237, Lys for the amino acid at position 330, Arg for the amino acid at position 330, Asp for the amino acid at position 233, Asp for the amino acid at position 268, Glu for the amino acid at position 268, Asp for the amino acid at position 326, Ser for the amino acid at position 326, Thr for the amino acid at position 326, Ile for the amino acid at position 323, Leu for the amino acid at position 323, Met for the amino acid at position 323, Asp for the amino acid at position 296, Ala for the amino acid at position 326, Asn for the amino acid at position 326, and Met for the amino acid at position 330, according to EU numbering.


Known methods such as site-directed mutagenesis (Kunkel et al. (Proc. Natl. Acad. Sci. USA (1985) 82, 488-492)) and Overlap extension PCR can be appropriately employed to alter the amino acids of Fc regions. Furthermore, various known methods can also be used as an amino acid alteration method for substituting amino acids by those other than natural amino acids (Annu Rev. Biophys. Biomol. Struct. (2006) 35: 225-249; Proc. Natl. Acad. Sci. U.S.A. (2003) 100(11): 6353-6357). For example, a cell-free translation system (Clover Direct (Protein Express)) containing tRNAs in which amber suppressor tRNA, which is complementary to UAG codon (amber codon) that is a stop codon, is linked with an unnatural amino acid may be suitably used.


In an embodiment of variants of the present invention, polynucleotides encoding antigen-binding molecules which have a heavy chain where a polynucleotide encoding an Fc region modified to have an amino acid mutation as described above is linked in frame to a polynucleotide encoding the above-described antigen-binding molecule whose binding activity varies depending on a selected condition.


The present invention provides methods for producing antigen-binding molecules, comprising collecting the antigen-binding molecules from culture media of cells introduced with vectors in which a polynucleotide encoding an Fc region is operably linked in frame to a polynucleotide encoding an antigen-binding domain whose binding activity varies depending on ion concentration condition. Furthermore, the present invention also provides methods for producing antigen-binding molecules, comprising collecting the antigen-binding molecules from culture media of cells introduced with vectors constructed by operably linking a polynucleotide encoding an antigen-binding domain whose binding activity varies depending on ion concentration condition to a polynucleotide encoding an Fc region which is in advance operably linked to a vector.


Pharmaceutical Composition

The present invention also relates to pharmaceutical compositions comprising antigen-binding molecules of the present invention, antigen-binding molecules produced by alteration methods of the present invention, or antigen-binding molecules produced by production methods of the present invention. Antigen-binding molecules of the present invention or antigen-binding molecules produced by production methods of the present invention are useful as pharmaceutical compositions since they, when administered, have the strong effect to reduce the plasma antigen concentration as compared to typical antigen-binding molecules, and exhibit the improved in vivo immune response, pharmacokinetics, and others in animals administered with the molecules. The pharmaceutical compositions of the present invention may comprise pharmaceutically acceptable carriers.


In the present invention, pharmaceutical compositions generally refer to 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. For example, they can be used parenterally, in the form of injections of sterile solutions or suspensions including water or other pharmaceutically acceptable liquid. For example, such compositions can be formulated by mixing in the form of unit dose required in the generally approved medicine manufacturing practice, by appropriately combining with pharmacologically acceptable carriers or media, specifically with sterile water, physiological saline, vegetable oil, emulsifier, suspension, surfactant, stabilizer, flavoring agent, excipient, vehicle, preservative, binder, or such. In such formulations, the amount of active ingredient is adjusted to obtain an appropriate amount in a pre-determined range.


Sterile compositions for injection can be formulated using vehicles such as distilled water for injection, according to standard formulation practice. 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). It is also possible to use in combination appropriate solubilizers, for example, alcohols (ethanol and such), polyalcohols (propylene glycol, polyethylene glycol, and such), non-ionic surfactants (polysorbate 80(TM), HCO-50, and such).


Oils include sesame oil and soybean oils. Benzyl benzoate and/or benzyl alcohol can be used in combination as solubilizers. It is also possible to combine 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. Appropriate ampules are filled with the prepared injections.


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


Administration methods can be appropriately selected in consideration of the patient's age and symptoms. The dose of a pharmaceutical composition containing an antigen-binding molecule can be, for example, from 0.0001 mg to 1000 mg/kg for each administration. Alternatively, the dose can be, for example, from 0.001 to 100000 mg per patient. However, the present invention is not limited by the numeric values described above. The doses and administration methods vary depending on the patient's weight, age, symptoms, and such. Those skilled in the art can set appropriate doses and administration methods in consideration of the factors described above.


Furthermore, the present invention provides kits for use in the methods of the present invention, which comprise at least an antigen-binding molecule of the present invention. In addition to the above, pharmaceutically acceptable carriers, media, instruction manuals describing the using method, and such may be packaged into the kits.


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.


Screening Method and Production Method

In an embodiment provided by the present invention, an antigen-binding molecule that binds to an aggregated antigen and has a function of eliminating the aggregated antigen from plasma may be obtained by screening antigen-binding molecules, which comprises the following step of:

  • (a) selecting an antigen-binding molecule whose antigen-binding activity to an aggregated antigen under an intracellular ion concentration condition is lower than the binding activity under an extracellular ion concentration condition.


In the screening method of the present invention, the above-mentioned ion concentration can be used as the ion concentration. For the intracellular ion concentration condition, for example, in the case of ionized calcium, the above-described low calcium concentration condition is applicable; and in the case of hydrogen ion or pH, the above-described high hydrogen ion concentration or low pH, i.e., an acidic pH range condition, is applicable. On the other hand, for the extracellular ion concentration, for example, the above-described high calcium concentration condition is applicable in the case of ionized calcium, and the above-described low hydrogen ion concentration or high pH, i.e., a neutral pH range condition is applicable in the case of hydrogen ion or pH. Whether the antigen-binding activity of the antigen-binding molecule is lower under an intracellular ion concentration condition than under an extracellular ion concentration condition can be confirmed by taking measurements under each of the ion concentration conditions according to known measurement methods such as those described in the above-described section “Binding Activity”.


The screening method of the present invention includes a method that further comprises the step(s) of:


(b):

  • (i) selecting an antigen-binding molecule whose binding activity to an aggregated antigen becomes higher than the binding activity to an unaggregated antigen under an extracellular ion concentration condition; and/or
  • (ii) selecting an antigen-binding molecule whose binding activity to an Fcγ receptor or an FcRn of a complex formed between an aggregated antigen and an antigen-binding molecule becomes higher than the binding activity to an Fcγ receptor or an FcRn of a complex formed between an unaggregated antigen and an antigen-binding molecule under an extracellular ion concentration condition.


    Carrying out these step(s) can yield an antigen-binding molecule that has a function of eliminating aggregated antigens in preference to unaggregated antigens.


Whether an antigen-binding molecule will show higher binding activity to an aggregated antigen than to an unaggregated antigen under an extracellular ion concentration condition can be confirmed by measuring its binding activity to the aggregated antigen and to the unaggregated antigen, respectively, under an extracellular ion concentration condition according to the above-described methods for measuring binding activity to an Fcγ receptor or FcRn.


Furthermore, “binding activity of an antigen-binding molecule to an aggregated antigen is higher than the binding activity to an unaggregated antigen under an extracellular ion concentration condition” can be reworded as “dissociation of an antigen-binding molecule from an aggregated antigen becomes slower than dissociation from an unaggregated antigen under an extracellular ion concentration condition”. Whether dissociation is becoming slow can be confirmed, for example, through comparison of the dissociation phases for the aggregated antigen and the unaggregated antigen, by using Biacore T200 (GE Healthcare) to obtain the slope of dissociation phase when the condition is changed from an extracellular ion concentration condition to an intracellular ion concentration condition. A specific method is as described in Example 2(2).


In one of the embodiments provided by the present invention, the antigen-binding molecule that binds to an aggregated antigen and has a function of eliminating the aggregated antigen from plasma may be obtained by a method of producing an antigen-binding molecule which comprises in addition to the above-described step (a) or steps (a) and (b), the following steps of:

  • (c) culturing a host cell comprising a vector carrying a gene encoding the antigen-binding molecule selected in step (a) or steps (a) and (b) mentioned above; and
  • (d) isolating an antigen-binding molecule from the culture obtained in step (c) mentioned above.


These steps can be carried out using a known antibody production method such as described in the above-mentioned section “Antibody”.


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









TABLE 7







LIST OF APPLICABLE DISEASES








DISEASE NAME
CAUSAL PROTEIN





Huntington('s) disease
huntingtin


Spinocerebellar ataxia type 1
ataxin-1


Spinocerebellar ataxia type 2
ataxin-2


Spinocerebellar ataxia type 6
Ca channel α1A


Spinocerebellar ataxia type 7
ataxin-7


Spinocerebellar ataxia type 17
TATA binding protein


Machado-Joseph disease
MDJ


Dentatorubropallidoluysian atrophy
DRPLA


Spinal and bulbar muscular atrophy
Androgen receptor


α1-antitrypsin deficiency
α1-antitrypsin


Emphysema
α1-antichymotrypsin


Premature senility
neuroserpin


Angioedema
C1 inhibitor


Generalized thrombus
Antithrombin III


Alzheimer('s) disease



AL amyloidosis
L-ch


Familial polyneuropathy (FAP)
trans thyretin


Reactive AA amyloidosis
SAA


Dialysis amyloidosis
β2M


AH amyloidosis
H-ch


Cerebral amyloid angiopathy
cystatin C


Parkinson('s) disease
α synuclein


Dementia with Lewy bodies
α synuclein


Multiple system degeneration
α synuclein


Type 2 diabetes
amylin


Sickle-cell anemia
hemoglobin


Cataract
crystallin


IgA nephropathy
IgA


Tauopathy
Tau protein


Frontotemporal lobar degeneration (FTLD)
TAR DNA-binding protein 43 kDa (TDP-



43)


Amyotrophic lateral sclerosis (ALS)
TAR DNA-binding protein 43 kDa (TDP-



43), Superoxide dismutase 1 (SOD1), FUS



(Fused in Sarcoma gene)


Creutzfeldt-Jakob disease: CJD
Prion


Gerstmann-Sträussler-Scheinker
Prion


syndrome: GSS


Fatal Familial Insomnia: FFI
Prion


Kuru
Prion


Charcot-Marie-Tooth disease: CMT1A
Prion


Congenital central hypoventilation
PHOX2B


syndrome


X-linked epilepsia nutans
ARX


Oculopharyngeal muscular dystrophy
Poly-adenylate binding protein nuclear 1



(PABPN1)


Limb-Girdle (distal/Miyoshi) muscular
dysferlin


dystrophy


Desmin myopathy
desmin


Leukodystrophies (Alexander disease)
GFAP


Epidermolysis bullosa Köbner (ichthyosis)
Keratin 5/14









EXAMPLES
Example 1
Preparation of Antibodies that Show Calcium-Dependent Binding to Human IgA
(1-1) Preparation of MRA-hIgA, GC-hIgA-FLAG, and GC-hIgA-MYC

As human IgA, MRA-hIgA (heavy chain SEQ ID NO: 33; light chain SEQ ID NO: 36),


GC-hIgA-FLAG (heavy chain SEQ ID NO: 34; light chain SEQ ID NO: 37), and GC-hIgA-MYC (heavy chain SEQ ID NO: 35; light chain SEQ ID NO: 37) were prepared as follows.


Preparation of MRA-hIgA

MRA-hIgA which is a recombinant of human IgA (hereinafter, MRA-hIgA) was prepared as follows. A gene fragment encoding MRA-hIgA (heavy chain SEQ ID NO: 33; light chain SEQ ID NO: 36) was inserted into an animal cell expression vector. The constructed plasmid vector was transfected into FreeStyle 293 (Invitrogen) using 293Fectin (Invitrogen) along with an EBNA1-expressing gene. Then, the transfected cells were cultured at 37° C. under 8% CO2 to secrete the MRA-IgA protein into the culture supernatant. The protein was purified using ion exchange chromatography and gel filtration chromatography according to a method known to those skilled in the art.


Preparation of GC-hIgA-FLAG

GC-hIgA-FLAG, which is a recombinant of human IgA, (hereinafter, GC-hIgA-FLAG) was prepared as follows. A gene fragment encoding GC-hIgA-FLAG (heavy chain SEQ ID NO: 34; light chain SEQ ID NO: 37) was inserted into an animal cell expression vector. The constructed plasmid vector was transfected into FreeStyle 293 (Invitrogen) using 293Fectin (Invitrogen) along with an EBNA1-expressing gene. Then, the transfected cells were cultured at 37° C. under 8% CO2 for six days to secrete the GC-hIgA protein into the culture supernatant.


The cell culture containing GC-hIgA-FLAG was filtered through a 0.22-μm bottle top filter to obtain culture supernatant. Purified GC-hIgA-FLAG was obtained using ion exchange chromatography and gel filtration chromatography according to a method known to those skilled in the art.


Preparation of GC-hIgA-MYC

GC-hIgA-MYC, which is a recombinant of human IgA, (hereinafter, GC-hIgA-MYC) was prepared as follows. A gene fragment encoding GC-hIgA-MYC (heavy chain SEQ ID NO: 35; light chain SEQ ID NO: 37) was inserted into an animal cell expression vector. The constructed plasmid vector was transfected into FreeStyle 293 (Invitrogen) using 293Fectin (Invitrogen) along with an EBNA1-expressing gene. Then, the transfected cells were cultured at 37° C. under 8% CO2 for six days to secrete the GC-hIgA protein into the culture supernatant.


The cell culture containing GC-hIgA-MYC was filtered through a 0.22-μm bottle top filter to obtain culture supernatant. Purified GC-hIgA-MYC was obtained using ion exchange chromatography and gel filtration chromatography according to a method known to those skilled in the art.


(1-2) Antibodies with Calcium-Dependent Binding

H54/L28-IgG1 described in International Publication No. WO 2009/125825 is a humanized anti-IL-6 receptor antibody. Fv4-IgG1 is a humanized anti-IL-6 receptor antibody that results from conferring H54/L28-IgG1 with the property of binding to soluble human IL-6 receptor in a pH-dependent manner (i.e., of binding under neutral condition and dissociating under acidic condition). The in vivo test described in International Publication No. WO 2009/125825 using mice demonstrated that elimination of soluble human IL-6 receptor is greatly accelerated in a group administered with a mixture of Fv4-IgG1 and soluble human IL-6 receptor as antigen as compared to a group administered with a mixture of H54/L28-IgG1 and soluble human IL-6 receptor as antigen.


Soluble human IL-6 receptor bound to a general antibody that binds to soluble human IL-6 receptor is recycled to the plasma along with the antibody via FcRn. Meanwhile, an antibody that binds to soluble human IL-6 receptor in a pH-dependent manner dissociates under the acidic conditions in the endosome from the soluble human IL-6 receptor that was bound to the antibody. The dissociated soluble human IL-6 receptor is degraded in the lysosome. Thus, this can greatly accelerate the elimination of soluble human IL-6 receptor. Moreover, the antibody that binds to soluble human IL-6 receptor in a pH-dependent manner is recycled to the plasma via FcRn, so that the recycled antibody can bind to a soluble human IL-6 receptor again. By repeating this, a single antibody molecule can repeatedly bind to soluble human IL-6 receptors multiple times (FIG. 1).


Meanwhile, as described in International Publication No. WO 2009/125825, H54/L28-IgG1 is a humanized anti-IL-6 receptor antibody and Fv4-IgG1 is a humanized anti-IL-6 receptor antibody that results from conferring H54/L28-IgG1 with the property of binding to soluble human IL-6 receptor in a pH-dependent manner (i.e., binding under neutral condition and dissociating under acidic condition). Fv4-IgG1-v2 is a humanized anti-IL-6 receptor antibody in which FcRn binding is increased over Fv4-IgG1 under neutral conditions. The in vivo test described in International Publication No. WO 2011/122011 using mice demonstrated that elimination of soluble human IL-6 receptor is greatly accelerated in a group administered with a mixture of Fv4-IgG1-v2 and soluble human IL-6 receptor as antigen as compared to a group administered with a mixture of Fv4-IgG1 and soluble human IL-6 receptor as antigen. Thus, it was reported that, by enhancing the binding toward FcRn under neutral condition (pH 7.4) of an antibody that binds to antigens in a pH-dependent manner, the effect of the antibody to repeatedly bind to antigens and the effect of promoting elimination of antigens from the plasma can be further improved, and antigen elimination can be eliminated from the plasma through administration of the antibody (FIG. 2).


In the mechanism of a pH-dependent binding antibody shown in FIGS. 1 and 2, it is important that the antibody strongly binds to an antigen in plasma and dissociates from the antigen in the endosome based on the environmental difference between plasma and endosome, i.e., pH difference (pH 7.4 in plasma; pH 6.0 in endosome). The degree of environmental difference between plasma and endosome is important for differentiating the antigen-binding ability of a pH-dependent binding antibody in plasma and endosome. A pH difference is due to a difference in the hydrogen ion concentration. Specifically, the hydrogen ion concentration in plasma (pH 7.4) is about 40 nM, while the concentration in the endosome (pH 6.0) is about 1000 nM. The factor (hydrogen ion) concentration differs by about 25 times between plasma and endosome.


The present inventors conceived that, in order to achieve the mechanism illustrated in FIGS. 1 and 2 easily or to enhance the mechanism, it would be beneficial to use an antibody that depends on a factor that has a greater concentration difference between plasma and endosome than the difference of hydrogen ion concentration between the two. Thus, the inventors searched for a factor whose concentration is considerably different between plasma and endosome. As a result, calcium was identified. The ionized calcium concentration is about 1.1 to 1.3 mM in plasma and about 3 μM in the endosome. The factor (calcium) concentration differs by about 400 times between the two. Thus, the ratio was found to be greater than the difference in hydrogen ion concentration (25 times). Specifically, antigen dissociation from an antibody which is equivalent to or more efficiently than a pH-dependent binding antibody is expected to be achieved by using an ionized calcium concentration-dependent binding antibody, which binds to an antigen under a high calcium concentration condition (1.1 to 1.3 mM) but dissociates from the antigen under a low calcium concentration condition (3 μM).


(1-3) Expression and Purification of Antibodies that Bind to Human IgA

GA2-IgG1 (heavy chain SEQ ID NO: 38; light chain SEQ ID NO: 39) is a newly obtained antibody that bind to human IgA. DNA sequence encoding GA2-IgG1 (heavy chain SEQ ID NO: 38; light chain SEQ ID NO: 39) were inserted into animal cell expression plasmids by a method known to those skilled in the art. Antibodies were expressed by the following method. Cells of human fetal kidney cell-derived FreeStyle 293-F (Invitrogen) were suspended in the FreeStyle 293 Expression Medium (Invitrogen), and plated at a cell density of 1.33×106 cells/ml (3 ml) into each well of a 6-well plate. The constructed plasmids were transfected into cells by a lipofection method. The cells were cultured for five days in a CO2 incubator (37° C., 8% CO2, 90 rpm). From the prepared culture supernatants, antibodies were purified using the rProtein A Sepharose™ Fast Flow (Amersham Biosciences) by a method known to those skilled in the art. The concentrations of purified antibodies were determined by measuring absorbance at 280 nm using a spectrophotometer. Antibody concentrations were calculated from the determined values using an extinction coefficient calculated by the PACE method (Protein Science (1995) 4: 2411-2423).


(1-4) Assessment of Prepared Antibodies for Calcium-Dependent Human IgA-Binding Activity

Using Biacore T200 (GE Healthcare), the obtained antibodies were assessed for their binding activity to human IgA (dissociation constant KD (M)). The measurement was carried out using as a running buffer 0.05% tween20, 20 mmol/l ACES, 150 mmol/1NaCl (pH 7.4 or pH 5.8) containing 3 μM or 1.2 mM CaCl2, or 0.05% tween20, 20 mmol/l ACES, 150 mmol/l NaCl (pH 8.0) containing 0.1 μM or 10 mM CaCl2.


After an adequate amount of recombinant Protein A/G (Thermo Scientific) was immobilized onto the Sensor chip CM5 (GE Healthcare) by an amino coupling method, antibodies were allowed to bind onto the sensor chip. An appropriate concentration of MRA-hIgA (described in (1-1)) was injected as an analyte to interact with antibodies on the sensor chip. Then, the sensor chip was regenerated by injecting 10 mmol/l glycine-HCl, pH 1.5. The measurement was carried out at 37° C. From the assay result, the dissociation constant KD (M) was calculated based on curve-fitting analysis and equilibrium constant analysis using Biacore T200 Evaluation Software (GE Healthcare). The result is shown in Table 8. It was revealed that GA2-IgG1 bound strongly to human IgA at a Ca2+ concentration of 1.2 mM whereas the antibody bound weakly to human IgA at a Ca2+ concentration of 3 Furthermore, under a 1.2 mM Ca2+ concentration condition, GA2-IgG1 was shown to bind to human IgA strongly at pH 7.4 but weakly at pH 5.8. More specifically, GA2-IgG1 was revealed to bind to human IgA in a pH- and calcium-dependent manner.














TABLE 8





Antibody







Name
Conditions
Fit
ka
kd
KD [M]







GA2-IgG1
pH 7.4, 1.2 mM Ca
1:1 binding model
4.0E+05
1.6E−02
3.9E−08



pH 7.4, 3 μM Ca
Steady State Affinity


6.7E−06



pH 5.8, 1.2 mM Ca
Steady State Affinity


4.0E−06



pH 5.8, 3 μM Ca
Steady State Affinity


5.0E−06









Example 2
(2-1) Preparation of Aggregated hIgA

Aggregated hIgA was prepared using the crosslinking agent SPDP (N-Succinimidyl 3-(2-pyridyldithio)propionate, Thermo Scientific). GC-hIgA-MYC prepared in Example 1 was modified using SPDP, and hIgAs were cross-linked with each other by mixing a fraction that has not been treated subsequently with a fraction that has been treated under reducing conditions. After the crosslinking reaction, the macromolecular component was fractioned by gel filtration chromatography to obtain aggregated hIgA. The result of gel filtration chromatographic analysis on aggregated hIgA is shown in FIG. 3. The apparent molecular weight of the aggregated hIgA, calculated from the elution position of the molecular-weight marker, was 780 kDa. Since the elution peak is broad, aggregates of various sizes are conceivably being formed. As a comparison, GC-hIgA-FLAG that has not been treated with SPDP was used as the unaggregated hIgA.


(2-2) Assessment of the Obtained Antibodies for their pH/Ca-Dependent Binding Ability to Aggregated hIgA

The obtained antibodies were assessed for their binding to unaggregated hIgA and aggregated hIgA using Biacore T200 (GE Healthcare). The measurement was carried out using as a running buffer, 0.05% tween20, 20 mmol/l ACES, 150 mmol/L NaCl (pH 7.4 and pH 5.8) containing 3 μM or 1.2 mM CaCl2.


After an adequate amount of recombinant Protein A/G (Thermo Scientific) was immobilized onto Sensor chip CM5 (GE Healthcare) by an amino coupling method, antibodies were allowed to bind onto the sensor chip. An appropriate concentration of hIgA (described in Example 1) was injected as an analyte to interact with antibodies on the sensor chip. Then, the sensor chip was regenerated by injecting 10 mmol/L Glycine-HCl, pH 1.5. The measurement was carried out at 37° C. The obtained sensorgram is shown in FIG. 4.


Binding of GA2-IgG1 to unaggregated hIgA was clearly shown to be strong at a Ca2+ concentration of 1.2 mM but weak at a Ca2+ concentration of 3 Comparison of the results from unaggregated hIgA and aggregated hIgA showed that the slope of the dissociation phase was becoming shallow for aggregated hIgA, and the dissociation of GA2-IgG1 was getting difficult. The reason for this delay of dissociation may be because a single aggregated hIgA molecule contains multiple binding sites and GA2-IgG1 binds to the molecule with avidity.


Example 3
Assessment of Human IgA-Binding Antibodies for their Effect on Plasma Retention of Aggregated hIgA and Unaggregated hIgA Using Normal Mice
(3-1) In Vivo Test Using Normal Mice

Normal mice (C57BL/6J mouse; Charles River Japan) were administered with unaggregated hIgA (prepared in Example 1) or aggregated hIgA (prepared in Example 2) alone or administered with an anti-hIgA antibody one day prior to administration of unaggregated hIgA or aggregated hIgA; and then they were assessed for the in vivo dynamics of hIgA and the anti-hIgA antibody. An anti-hIgA antibody, an unaggregated hIgA solution (300 μh/mL) and an aggregated hIgA solution (300 μg/mL) were administered at 10 mL/kg into the caudal vein. The anti-hIgA antibody used was GA2-IgG1 described above.


The concentration of human IgA was 300 μg/mL in all of the mixed solutions. Meanwhile, the anti-hIgA antibody concentration was 30 μg/mL (0.3 mg/kg), 100 μg/mL (1 mg/kg), or 300 μg/mL (3 mg/kg). When the anti-hIgA antibody concentration was 30 μg/mL or 100 μg/mL, hIgA was present in excess relative to the anti-hIgA antibody, and therefore it was assumed that majority of the anti-hIgA antibody was bound to hIgA. Conversely, more than half of hIgA was not bound with the antibody. Meanwhile, when the anti-hIgA antibody concentration was 300 μg/mL, the anti-hIgA antibody and hIgA were present in nearly equal amounts; and when the affinity of GA2-IgG1 was taken into account, 80% or so of both the anti-hIgA antibody and hIgA were thought to be bound.


Blood was collected 5 minutes, 1 hour, 2 hours, 4 hours, 7 hours, 1 day, 2 days, and 3 days, after administration. Immediately after the collection, the blood was centrifuged at 4° C. and 12000 rpm for 15 minutes to isolate plasma. The isolated plasma was stored in a freezer at −20° C. or below before measurements.


(3-2) Determination of Plasma Concentration of Anti-Human IgA Antibody in Normal Mice by ELISA

Anti-human IgA antibody concentrations in mouse plasma were determined by ELISA. First, Anti-Human IgG (γ-chain specific) F(ab′)2 Fragment of Antibody (SIGMA) was aliquoted into Nunc-Immuno Plate, MaxiSorp (Nalge nunc International). The plate was left overnight at 4° C. to prepare an anti-human IgG antibody-immobilized plate. Standard samples were prepared at plasma concentrations of 2, 1, 0.5, 0.25, 0.125, 0.0625, and 0.03125 μg/ml. Mouse plasma assay samples were prepared by diluting 100 times or more. The samples were aliquoted into the Anti-Human IgG antibody-immobilized plate. After one hour of incubation at room temperature, the samples were reacted with the Goat Anti-Human IgG (γ chain specific) Biotin (BIOT) Conjugate (Southern Biotechnology Associats Inc.) at room temperature for one hour. Then, the samples were reacted with Streptavidin-PolyHRP80 (Stereospecific Detection Technologies) at room temperature for one hour. Chromogenic reaction was carried out using the TMB One Component HRP Microwell Substrate (BioFX Laboratories) as a substrate. After the reaction was terminated with 1N sulfuric acid (Showa Chemical), the absorbance at 450 nm was measured using a microplate reader. Using the analysis software SOFTmax PRO (Molecular Devices), the concentrations in mouse plasma were calculated based on the absorbance from the standard curve. A time course of plasma concentrations of antibody GA2-IgG1 determined by the above-described method after intravenous administration to normal mice is shown in FIG. 5.


(3-3) Determination of Plasma Human IgA Concentration by ELISA

The concentration of unaggregated human IgA (monomeric human IgA) in mouse plasma was determined by ELISA. First, the Mouse anti-FLAG Antibody (SIGMA) was dispensed into the Nunc-Immuno Plates, MaxiSorp (Nalge nunc International), and allowed to stand overnight at 4° C. to prepare Anti-FLAG-immobilized plates. Human IgA calibration curve samples of plasma concentrations 20, 10, 5, 2.5, 1.25, 0.625, and 0.3125 μg/mL, and mouse plasma assay samples diluted 100-fold or more were prepared and aliquoted into the Anti-FLAG-immobilized plates. The plates were incubated overnight at room temperature. 100 μL of 500 ng/mL GPC3 (R&D Systems) was added and reacted at room temperature for one hour. 100 μL of 1 μg/mL Anti-GPC3 Antibody Biotinylated was added and reacted at room temperature for one hour, and then with the Streptavidin-Po1yHRP80 (Stereospecific Detection Technologies) at room temperature for one hour. Chromogenic reaction was carried out using the TMB One Component HRP Microwell Substrate (BioFX Laboratories) as a substrate. After the reaction was terminated with 1N sulfuric acid (Showa Chemical), the absorbance at 450 nm was measured using a microplate reader. Using analysis software SOFTmax PRO (Molecular Devices), the concentrations in mouse plasma were calculated based on the absorbance from the standard curve.


The human IgA-SPDP-Polymer (aggregated human IgA) concentration in mouse plasma was measured by the ECL method. First, a Mouse Anti-c-MYC Antibody (SIGMA) was dispensed into the MULTI-ARRAY-96-well Plate (MSD), and allowed to stand for one hour at room temperature to prepare Anti-MYC-immobilized plates. Human IgA-SPDP-Polymer calibration curve samples with plasma concentrations of 10, 5, 2.5, 1.25, 0.625, 0.3125, and 0.1563 μg/mL, and mouse plasma assay samples diluted 100-fold or more were prepared. In preparation, first, mouse plasma assay samples and 50-fold-diluted plasma containing STD were prepared using a buffer containing 10 mM EDTA, and then this was mixed with an equal amount of 20 μg/mL antibody to be administered (in 10 mM EDTA buffer) and incubated at room temperature for 30 minutes to prepare samples for addition to plates. These samples were aliquoted into the Anti-MYC-immobilized plate and then incubated at room temperature for one hour. Subsequently, 100 μL of 1 μg/mL Rabbit Anti-human-IgA-Antibody Biotinylated (BETHYL) was added and reacted at room temperature for one hour, and this was further reacted with Streptavidin-PolyHRP80 (Stereospecific Detection Technologies) at room temperature for one hour. Next, after addition of Read Buffer, measurements were taken on Sector Imager 2400 (MSD). The concentration in mouse plasma was calculated based on the response in the calibration curve using the analytical software, SOFTmax PRO (Molecular Devices). A time course of human IgA (unaggregated human IgA (monomeric human IgA)) concentration and human IgA-SPDP-Polymer (aggregated human IgA) concentration in normal mouse plasma after intraveous administration, which were measured by this method, are shown in FIGS. 6 and 7, respectively, and their clearance values and such are shown in Table 9.













TABLE 9









Clearance ratio





Clearance ratio
(aggregated human




Human IgA
CL (human IgA +
IgA)/clearance



GA2-IgG1 dose
clearance
GA2-IgG1)/CL
rate (monomeric


Human IgA
(mg/kg)
(mL/day/kg)
(human IgA alone)
human IgA)







Monomeric

80.6 ± 5.2




human IgA
0.3
 85.5 ± 20.6
1.1




1.0
98.5 ± 5.3
1.2




3.0
128 ± 6 
1.6



Aggregated

307 ± 19




human IgA
0.3
557 ± 42
1.8
1.7



1.0
1599 ± 359
5.2
4



3.0
4967 ± 411
16.2 
10.2









Time course of “concentration of unaggregated human IgA after GA2-IgG1 administration/concentration of unaggregated human IgA when human IgA alone is administered” and “concentration of aggregated human IgA after GA2-IgG1 administration/concentration of aggregated human IgA when human IgA alone is administered” are shown in FIG. 8 as indicators that present the degree at which elimination of both types of human IgA due to administration of GA2-IgG1 was accelerated as compared to administration of monomeric human IgA (unaggregated human IgA) alone or aggregated human IgA alone.


These results show that at any dose, GA2-IgG1 accelerates elimination of aggregated human IgA in preference to unaggregated human IgA. In particular in GA2-IgG1 at 3 mg/kg, clearance of aggregated human IgA could be accelerated preferentially by ten times or more in terms of clearance ratio.


DISCUSSION

As a result, while clearance of unaggregated human IgA was only slightly faster when co-administered with GA2-IgG1 than when administered alone, clearance of aggregated human IgA was greatly accelerated when co-administered with GA2-IgG1 as compared to when administered alone. While the clearance ratio of unaggregated human IgA between single-administration and co-administration with an antibody is 1.1 to 1.6, the clearance ratio of aggregated human IgA between single administration and co-administration with an antibody is 1.8 to 16.2. This shows that the degree of acceleration of clearance is greater for aggregated human IgA than for unaggregated human IgA when co-administered with an antibody.


Regarding the pharmacokinetics of GA2-IgG1, also when it was co-administered with either unaggregated human IgA or aggregated human IgA, elimination was slow one day after administration and onwards.


The following mechanism may be the reason why clearance of aggregated human IgA was greatly accelerated as compared to clearance of unaggregated human IgA.


Since an aggregated antigen contains multiple antibody-binding sites in a single molecule, multiple antibody molecules are polyvalently and strongly bound; and it hardly dissociates compared to the unaggregated monomeric antigen, and forms huge immune complexes. As shown in FIG. 9, since huge immune complexes containing multiple antibody molecules can bind polyvalently and strongly to cell surface receptors (FcγR, FcRn, complement receptors, and such) via a polyvalent Fc region, they are efficiently taken up by cells expressing these receptors. Then, dissociation of the aggregated antigen from the antibody showing pH- or Ca-dependent binding dissolves the formation of immune complexes in the endosome. Since the aggregated antigen cannot bind to FcRn, it is transferred to a lysosome and then degraded. Meanwhile, since the antibody cannot form an immune complex, it is thought to be recycled into plasma by FcRn. In contrast, since small immune complexes not containing multiple antibodies do not have a sufficient affinity to natural IgG1-type receptors as shown in FIG. 10, their incorporation into cells is low in efficiency or takes place only non-specifically.


As shown in FIG. 4, when GA2-IgG1 is allowed to interact with aggregated human IgA, the dissociation phase becomes shallow; and compared to dissociation from unaggregated human IgA, GA2-IgG1 has the property of not being easily dissociated from aggregated human IgA. More specifically, since multiple GA2-IgG1 molecules bind multivalently to an aggregated human IgA and dissociation is difficult, they may form a larger immune complex than the immune complex formed between unaggregated human IgA and GA2-IgG1. Since huge immune complexes bind strongly via multivalent Fc regions to FcγR, FcRn, complement receptors, or such, it is thought that they are quickly taken up by cells expressing these receptors, and then the aggregated human IgA dissociates from GA2-IgG1 in the endosome and is transferred to the lysosome and degraded, and thus the clearance was substantially accelerated compared to when human IgA was administered alone. Furthermore, GA2-IgG1 is considered to have been recycled into plasma by FcRn after dissociation of aggregated human IgA and dissolution of the immune complex.


More specifically, GA2-IgG1 may have accomplished substantial acceleration of clearance of aggregated human IgA by two multivalent-binding effects: forming a huge immune complex by strongly and multivalently binding to aggregated human IgA more than to unaggregated human IgA, and strongly and multivalently binding to various Fc receptors via multiple Fe's.


As such, it was shown that GA2-IgG1 has the effect of eliminating aggregated human IgA from plasma in preference to unaggregated human IgA.


Therefore, clearance of aggregated antigens can be preferentially accelerated in the co-presence of unaggregated and aggregated antigens, by conferring an antigen-binding molecule that binds to aggregated antigens and comprises an Fc region with the function of having a pH- and/or Ca-concentration-dependent binding activity. Furthermore, it may be possible to increase preference for aggregated antigens and accelerate clearance of aggregated antigens if an appropriate pH- and Ca-dependent antibody that binds to aggregated antigens more strongly than to unaggregated antigens can be selected.


Antigen-binding molecules that have an effect of eliminating aggregated antigens in preference to unaggregated antigens from plasma in this manner may be applied as therapeutic agents for diseases caused by aggregates such as amyloidosis, polyglutamine disease, serpin disease, IgA nephropathy, and such.

Claims
  • 1.-34. (canceled)
  • 35. An antigen-binding molecule comprising an antigen-binding domain and an Fc region, wherein the antigen-binding domain binds to an aggregated form of an antigen with a binding strength that varies depending on pH, calcium ion concentration, or both; and wherein the antigen naturally aggregates in plasma in vivo to produce the aggregated form.
  • 36. The antigen-binding molecule of claim 35, wherein the antigen-binding molecule binds more strongly to the aggregated form of the antigen than to an unaggregated form of the antigen.
  • 37. The antigen-binding molecule of claim 35, wherein a first complex comprising the antigen-binding molecule and the aggregated form of the antigen binds to a human Fcγ or FcRn receptor more strongly than does a second complex comprising the antigen-binding molecule and an unaggregated form of the antigen.
  • 38. The antigen-binding molecule of claim 36, wherein, when the antigen-binding molecule is introduced into plasma containing both the aggregated and unaggregated forms of the antigen, the antigen-binding molecule removes the aggregated form from the plasma in preference to the unaggregated form.
  • 39. The antigen-binding molecule of claim 38, wherein the ratio of plasma clearance of the aggregated form of the antigen in the presence of the antigen-binding molecule to plasma clearance of the aggregated form of the antigen in the absence of the antigen-binding molecule is at least 1.5 times the ratio of plasma clearance of the unaggregated form of the antigen in the presence of the antigen-binding molecule to plasma clearance of the unaggregated form of the antigen in the absence of the antigen-binding molecule.
  • 40. The antigen-binding molecule of claim 35, wherein the binding strength varies depending on calcium ion concentration.
  • 41. The antigen-binding molecule of claim 40, wherein the binding strength is lower at a first calcium concentration that is between 1 μM and 5 μM than at a second calcium concentration that is between 500 μM and 2.5 mM.
  • 42. The antigen-binding molecule of claim 35, wherein the binding strength varies depending on pH.
  • 43. The antigen-binding molecule of claim 42, wherein the binding strength is lower at pH 5.8 than at pH 7.4.
  • 44. The antigen-binding molecule of claim 35, wherein the antigen is selected from the group consisting of huntingtin, ataxin-1, ataxin-2, Ca channel α1A, ataxin-7, TATA binding protein, Machado-Joseph disease protein (MJD), Dentatorubropallidoluysian atrophy protein (DRPLA), androgen receptor, α1-antitrypsin, α1-antichymotrypsin, neuroserpin, C1 inhibitor, antithrombin III, amyloid-β (Aβ), immunoglobulin L chain (L-ch), transthyretin, serum amyloid A (SAA), β2 microglobulin (β2M), immunoglobulin H chain (H-ch), cystatin C, a synuclein, amylin, hemoglobin, crystalline, immunoglobulin A (IgA), Tau protein, TAR DNA-binding protein 43 kDa (TDP-43), Superoxide dismutase (SOD1), Fused in Sarcoma gene (FUS), prions, Paired-like homeobox 2b (PHOX2B), aristaless related homeobox (ARX), poly-adenylate binding protein nuclear 1 (PABPN1), dysferlin, desmin, Glial fibrillary acidic protein (GFAP), and keratin 5/14.
  • 45. The antigen-binding molecule of claim 35, wherein the Fc region is a native human IgG Fc region comprising the amino acid sequence of SEQ ID NO: 9, 10, 11, or 12.
  • 46. The antigen-binding molecule of claim 35, wherein the Fc region is a modified Fc region whose FcRn-binding activity at pH 5.8 is increased compared to the FcRn-binding activity of a native human IgG Fc region at pH 5.8, wherein the native human IgG Fc region comprises SEQ ID NO: 9, 10, 11, or 12.
  • 47. The antigen-binding molecule of claim 46, wherein the modified Fc region differs from the native human IgG Fc region by amino acid substitution at one or more of the following positions (by EU numbering): 238, 244, 245, 249, 250, 251, 252, 253, 254, 255, 256, 257, 258, 260, 262, 265, 270, 272, 279, 283, 285, 286, 288, 293, 303, 305, 307, 308, 309, 311, 312, 314, 316, 317, 318, 332, 339, 340, 341, 343, 356, 360, 362, 375, 376, 377, 378, 380, 382, 385, 386, 387, 388, 389, 400, 413, 415, 423, 424, 427, 428, 430, 431, 433, 434, 435, 436, 438, 439, 440, 442, and 447.
  • 48. The antigen-binding molecule of claim 47, wherein at least one of the following positions (EU numbering) in the modified Fc region is occupied by the indicated amino acid: Leu at position 238;Leu at position 244;Arg at position 245;Pro at position 249;either Gln or Glu at position 250;any one of Arg, Asp, Glu, or Leu at position 251;any one of Phe, Ser, Thr, or Tyr at position 252;either Ser or Thr at position 254;any one of Arg, Gly, Ile, or Leu at position 255;any one of Ala, Arg, Asn, Asp, Gln, Glu, Pro, or Thr at position 256;any one of Ala, Ile, Met, Asn, Ser, or Val at position 257;Asp at position 258;Ser at position 260;Leu at position 262;Lys at position 270;either Leu or Arg at position 272;any one of Ala, Asp, Gly, His, Met, Asn, Gln, Arg, Ser, Thr, Trp, or Tyr at position 279;any one of Ala, Asp, Phe, Gly, His, Ile, Lys, Leu, Asn, Pro, Gln, Arg, Ser, Thr, Trp, or Tyr at position 283;Asn at position 285;Phe at position 286;either Asn or Pro at position 288;Val at position 293;any one of Ala, Glu, Gln, or Met at position 307;any one of Ala, Glu, Ile, Lys, Leu, Met, Ser, Val, or Trp at position 311;Pro at position 309;any one of Ala, Asp, or Pro at position 312;either Ala or Leu at position 314;Lys at position 316;Pro at position 317;either Asn or Thr at position 318;any one of Phe, His, Lys, Leu, Met, Arg, Ser, or Trp at position 332;any one of Asn, Thr, or Trp at position 339;Pro at position 341;any one of Glu, His, Lys, Gln, Arg, Thr, or Tyr at position 343;Arg at position 375;any one of Gly, Ile, Met, Pro, Thr, or Val at position 376;Lys at position 377;any one of Asp, Asn, or Val at position 378;any one of Ala, Asn, Ser, or Thr at position 380;any one of Phe, His, Ile, Lys, Leu, Met, Asn, Gln, Arg, Ser, Thr, Val, Trp, or Tyr at position 382;any one of Ala, Arg, Asp, Gly, His, Lys, Ser, or Thr at position 385;any one of Arg, Asp, Ile, Lys, Met, Pro, Ser, or Thr at position 386;any one of Ala, Arg, His, Pro, Ser, or Thr at position 387;any one of Asn, Pro, or Ser at position 389;Asn at position 423;Asn at position 427;any one of Leu, Met, Phe, Ser, or Thr at position 428;any one of Ala, Phe, Gly, His, Ile, Lys, Leu, Met, Asn, Gln, Arg, Ser, Thr, Val, or Tyr at position 430;either His or Asn at position 431;any one of Arg, Gln, His, Ile, Lys, Pro, or Ser at position 433;any one of Ala, Gly, His, Phe, Ser, Trp, or Tyr at position 434;any one of Arg, Asn, His, Ile, Leu, Lys, Met, or Thr at position 436;any one of Lys, Leu, Thr, or Trp at position 438;Lys at position 440, orLys at position 442; andany one of Ile, Pro, or Thr at position 308.
  • 49. The antigen-binding molecule of claim 35, wherein the Fc region is a modified Fc region whose FcRn-binding activity at pH 7.4 is increased compared to the FcRn-binding activity of a native human IgG Fc region at pH 7.4, wherein the native human IgG Fc region comprises SEQ ID NO: 9, 10, 11, or 12.
  • 50. The antigen-binding molecule of claim 49, wherein the modified Fc region differs from the native human IgG Fc region by amino acid substitution at one or more of the following positions (by EU numbering): 237, 248, 250, 252, 254, 255, 256, 257, 258, 265, 286, 289, 297, 298, 303, 305, 307, 308, 309, 311, 312, 314, 315, 317, 332, 334, 360, 376, 380, 382, 384, 385, 386, 387, 389, 424, 428, 433, 434, and 436.
  • 51. The antigen-binding molecule of claim 50, wherein at least one of the following positions (EU numbering) in the modified Fc region is occupied by the indicated amino acid: Met at position 237;Ile at position 248;any one of Ala, Phe, Ile, Met, Gln, Ser, Val, Tip, or Tyr at position 250;any one of Phe, Tip, or Tyr at position 252;Thr at position 254;Glu at position 255;any one of Asp, Asn, Glu, or Gln at position 256;any one of Ala, Gly, Ile, Leu, Met, Asn, Ser, Thr, or Val at position 257;His at position 258;Ala at position 265;either Ala or Glu at position 286;His at position 289;Ala at position 297;Ala at position 303;Ala at position 305;any one of Ala, Asp, Phe, Gly, His, Ile, Lys, Leu, Met, Asn, Pro, Gln, Arg, Ser, Val, Trp, or Tyr at position 307;any one of Ala, Phe, Ile, Leu, Met, Pro, Gln, or Thr at position 308;any one of Ala, Asp, Glu, Pro, or Arg at position 309;any one of Ala, His, or Ile at position 311;either Ala or His at position 312;either Lys or Arg at position 314;any one of Ala, Asp, or His at position 315;Ala at position 317;Val at position 332;Leu at position 334;His at position 360;Ala at position 376;Ala at position 380;Ala at position 382;Ala at position 384;either Asp or His at position 385;Pro at position 386;Glu at position 387;either Ala or Ser at position 389;Ala at position 424;any one of Ala, Asp, Phe, Gly, His, Ile, Lys, Leu, Asn, Pro, Gln, Ser, Thr, Val, Trp, or Tyr at position 428;Lys at position 433;any one of Ala, Phe, His, Ser, Trp, or Tyr at position 434;any one of His, Ile, Leu, Phe, Thr, or Val at position 436.
  • 52. The antigen-binding molecule of claim 35, wherein the Fc region is a modified Fc region whose Fcγ receptor-binding activity is increased compared to the Fcγ receptor-binding activity of a native human IgG Fc region.
  • 53. The antigen-binding molecule of claim 52, wherein the modified Fc region differs from the native human IgG Fc region at one or more of the following positions (by EU numbering): 221, 222, 223, 224, 225, 227, 228, 230, 231, 232, 233, 234, 235, 236, 237, 238, 239, 240, 241, 243, 244, 245, 246, 247, 249, 250, 251, 254, 255, 256, 258, 260, 262, 263, 264, 265, 266, 267, 268, 269, 270, 271, 272, 273, 274, 275, 276, 278, 279, 280, 281, 282, 283, 284, 285, 286, 288, 290, 291, 292, 293, 294, 295, 296, 297, 298, 299, 300, 301, 302, 303, 304, 305, 311, 313, 315, 317, 318, 320, 322, 323, 324, 325, 326, 327, 328, 329, 330, 331, 332, 333, 334, 335, 336, 337, 339, 376, 377, 378, 379, 380, 382, 385, 392, 396, 421, 427, 428, 429, 434, 436, and 440.
  • 54. The antigen-binding molecule of claim 53, wherein at least one of the following positions (EU numbering) in the modified Fc region is occupied by the indicated amino acid: either Lys or Tyr at position 221;any one of Phe, Trp, Glu, or Tyr at position 222;any one of Phe, Trp, Glu, or Lys at position 223;any one of Phe, Trp, Glu, or Tyr at position 224;any one of Glu, Lys, or Tip at position 225;any one of Glu, Gly, Lys, or Tyr at position 227;any one of Glu, Gly, Lys, or Tyr at position 228;any one of Ala, Glu, Gly, or Tyr at position 230;any one of Glu, Gly, Lys, Pro, or Tyr at position 231;any one of Glu, Gly, Lys, or Tyr at position 232;any one of Ala, Asp, Phe, Gly, His, Ile, Lys, Leu, Met, Asn, Gln, Arg, Ser, Thr, Val, Trp, or Tyr at position 233;any one of Ala, Asp, Glu, Phe, Gly, His, Ile, Lys, Met, Asn, Pro, Gln, Arg, Ser, Thr, Val, Trp, or Tyr at position 234;any one of Ala, Asp, Glu, Phe, Gly, His, Ile, Lys, Met, Asn, Pro, Gln, Arg, Ser, Thr, Val, Trp, or Tyr at position 235;any one of Ala, Asp, Glu, Phe, His, Ile, Lys, Leu, Met, Asn, Pro, Gln, Arg, Ser, Thr, Val, Trp, or Tyr at position 236;any one of Asp, Glu, Phe, His, Ile, Lys, Leu, Met, Asn, Pro, Gln, Arg, Ser, Thr, Val, Trp, or Tyr at position 237;any one of Asp, Glu, Phe, Gly, His, Ile, Lys, Leu, Met, Asn, Gln, Arg, Ser, Thr, Val, Trp, or Tyr at position 238;any one of Asp, Glu, Phe, Gly, His, Ile, Lys, Leu, Met, Asn, Pro, Gln, Arg, Thr, Val, Trp, or Tyr at position 239;any one of Ala, Ile, Met, or Thr at position 240;any one of Asp, Glu, Leu, Arg, Trp, or Tyr at position 241;any one of Leu, Glu, Leu, Gln, Arg, Trp, or Tyr at position 243;His at position 244;Ala at position 245;any one of Asp, Glu, His, or Tyr at position 246;any one of Ala, Phe, Gly, His, Ile, Leu, Met, Thr, Val, or Tyr at position 247;any one of Glu, His, Gln, or Tyr at position 249;either Glu or Gln at position 250;Phe at position 251;any one of Phe, Met, or Tyr at position 254;any one of Glu, Leu, or Tyr at position 255;any one of Ala, Met, or Pro at position 256;any one of Asp, Glu, His, Ser, or Tyr at position 258;any one of Asp, Glu, His, or Tyr at position 260;any one of Ala, Glu, Phe, Ile, or Thr at position 262;any one of Ala, Ile, Met, or Thr at position 263;any one of Asp, Glu, Phe, Gly, His, Ile, Lys, Leu, Met, Asn, Pro, Gln, Arg, Ser, Thr, Trp, or Tyr at position 264;any one of Ala, Leu, Phe, Gly, His, Ile, Lys, Leu, Met, Asn, Pro, Gln, Arg, Ser, Thr, Val, Trp, or Tyr at position 265;any one of Ala, Ile, Met, or Thr at position 266;any one of Asp, Glu, Phe, His, Ile, Lys, Leu, Met, Asn, Pro, Gln, Arg, Thr, Val, Trp, or Tyr at position 267;any one of Asp, Glu, Phe, Gly, Ile, Lys, Leu, Met, Pro, Gln, Arg, Thr, Val, or Trp at position 268;any one of Phe, Gly, His, Ile, Lys, Leu, Met, Asn, Pro, Arg, Ser, Thr, Val, Trp, or Tyr at position 269;any one of Glu, Phe, Gly, His, Ile, Leu, Met, Pro, Gln, Arg, Ser, Thr, Trp, or Tyr at position 270;any one of Ala, Asp, Glu, Phe, Gly, His, Ile, Lys, Leu, Met, Asn, Gln, Arg, Ser, Thr, Val, Trp, or Tyr at position 271;any one of Asp, Phe, Gly, His, Ile, Lys, Leu, Met, Pro, Arg, Ser, Thr, Val, Trp, or Tyr at position 272;either Phe or Ile at position 273;any one of Asp, Glu, Phe, Gly, His, Ile, Leu, Met, Asn, Pro, Arg, Ser, Thr, Val, Trp, or Tyr at position 274;either Leu or Trp at position 275;any one of Asp, Glu, Phe, Gly, His, Ile, Leu, Met, Pro, Arg, Ser, Thr, Val, Trp, or Tyr at position 276;any one of Asp, Glu, Gly, His, Ile, Lys, Leu, Met, Asn, Pro, Gln, Arg, Ser, Thr, Val, or Trp at position 278;Ala at position 279;any one of Ala, Gly, His, Lys, Leu, Pro, Gln, Trp, or Tyr at position 280;any one of Asp, Lys, Pro, or Tyr at position 281;any one of Glu, Gly, Lys, Pro, or Tyr at position 282;any one of Ala, Gly, His, Ile, Lys, Leu, Met, Pro, Arg, or Tyr at position 283;any one of Asp, Glu, Leu, Asn, Thr, or Tyr at position 284;any one of Asp, Glu, Lys, Gln, Trp, or Tyr at position 285;any one of Glu, Gly, Pro, or Tyr at position 286;any one of Asn, Asp, Glu, or Tyr at position 288;any one of Asp, Gly, His, Leu, Asn, Ser, Thr, Trp, or Tyr at position 290;any one of Asp, Glu, Gly, His, Ile, Gln, or Thr at position 291;any one of Ala, Asp, Glu, Pro, Thr, or Tyr at position 292;any one of Phe, Gly, His, Ile, Leu, Met, Asn, Pro, Arg, Ser, Thr, Val, Trp, or Tyr at position 293;any one of Phe, Gly, His, Ile, Lys, Leu, Met, Asn, Pro, Arg, Ser, Thr, Val, Trp, or Tyr at position 294;any one of Asp, Glu, Phe, Gly, His, Ile, Lys, Met, Asn, Pro, Arg, Ser, Thr, Val, Trp, or Tyr at position 295;any one of Ala, Asp, Glu, Gly, His, Ile, Lys, Leu, Met, Asn, Gln, Arg, Ser, Thr, or Val at position 296;any one of Asp, Glu, Phe, Gly, His, Ile, Lys, Leu, Met, Pro, Gln, Arg, Ser, Thr, Val, Trp, or Tyr at position 297;any one of Ala, Asp, Glu, Phe, His, Ile, Lys, Met, Asn, Gln, Arg, Thr, Val, Trp, or Tyr at position 298;any one of Ala, Asp, Glu, Phe, Gly, His, Ile, Lys, Leu, Met, Asn, Pro, Gln, Arg, Ser, Val, Trp, or Tyr at position 299;any one of Ala, Asp, Glu, Gly, His, Ile, Lys, Leu, Met, Asn, Pro, Gln, Arg, Ser, Thr, Val, or Trp at position 300;any one of Asp, Glu, His, or Tyr at position 301;Ile at position 302;any one of Asp, Gly, or Tyr at position 303;any one of Asp, His, Leu, Asn, or Thr at position 304;any one of Glu, Ile, Thr, or Tyr at position 305;any one of Ala, Asp, Asn, Thr, Val, or Tyr at position 311;Phe at position 313;Leu at position 315;either Glu or Gln at position 317;any one of His, Leu, Asn, Pro, Gln, Arg, Thr, Val, or Tyr at position 318;any one of Asp, Phe, Gly, His, Ile, Leu, Asn, Pro, Ser, Thr, Val, Trp, or Tyr at position 320;any one of Ala, Asp, Phe, Gly, His, Ile, Pro, Ser, Thr, Val, Trp, or Tyr at position 322;Ile at position 323;any one of Asp, Phe, Gly, His, Ile, Leu, Met, Pro, Arg, Thr, Val, Trp, or Tyr at position 324;any one of Ala, Asp, Glu, Phe, Gly, His, Ile, Lys, Leu, Met, Pro, Gln, Arg, Ser, Thr, Val, Trp, or Tyr at position 325;any one of Ala, Asp, Glu, Gly, Ile, Leu, Met, Asn, Pro, Gln, Ser, Thr, Val, Trp, or Tyr at position 326;any one of Ala, Asp, Glu, Phe, Gly, His, Ile, Lys, Leu, Met, Asn, Pro, Arg, Thr, Val, Trp, or Tyr at position 327;any one of Ala, Asp, Glu, Phe, Gly, His, Ile, Lys, Met, Asn, Pro, Gln, Arg, Ser, Thr, Val, Trp, or Tyr at position 328;any one of Asp, Glu, Phe, Gly, His, Ile, Lys, Leu, Met, Asn, Gln, Arg, Ser, Thr, Val, Trp, or Tyr at position 329;any one of Cys, Glu, Phe, Gly, His, Ile, Lys, Leu, Met, Asn, Pro, Arg, Ser, Thr, Val, Trp, or Tyr at position 330;any one of Asp, Phe, His, Ile, Leu, Met, Gln, Arg, Thr, Val, Trp, or Tyr at position 331;any one of Ala, Asp, Glu, Phe, Gly, His, Lys, Leu, Met, Asn, Pro, Gln, Arg, Ser, Thr, Val, Trp, or Tyr at position 332;any one of Ala, Asp, Glu, Phe, Gly, His, Ile, Leu, Met, Pro, Ser, Thr, Val, or Tyr at position 333;any one of Ala, Glu, Phe, Ile, Leu, Pro, or Thr at position 334;any one of Asp, Phe, Gly, His, Ile, Leu, Met, Asn, Pro, Arg, Ser, Val, Trp, or Tyr at position 335;any one of Glu, Lys, or Tyr at position 336;any one of Glu, His, or Asn at position 337;any one of Asp, Phe, Gly, Ile, Lys, Met, Asn, Gln, Arg, Ser, or Thr at position 339;either Ala or Val at position 376;either Gly or Lys at position 377;Asp at position 378;Asn at position 379;any one of Ala, Asn, or Ser at position 380;either Ala or Ile at position 382;Glu at position 385;Thr at position 392;Leu at position 396;Lys at position 421;Asn at position 427;either Phe or Leu at position 428;Met at position 429;Trp at position 434;Ile at position 436; andany one of Gly, His, Ile, Leu, or Tyr at position 440.
  • 55. The antigen-binding molecule of claim 35, wherein the Fc region binds more strongly to an inhibitory Fcγ receptor than to an activating Fcγ receptor.
  • 56. The antigen-binding molecule of claim 55, wherein the inhibitory Fcγ receptor is human FcγRIIb.
  • 57. The antigen-binding molecule of claim 55, wherein the activating Fcγreceptor is human FcγRIa, human FcγRIIa (R), human FcγRIIa (H), human FcγRIIIa (V), or human FcγRIIIa (F).
  • 58. The antigen-binding molecule of claim 55, wherein the Fc region is a modified Fc region that differs from a native human IgG Fc region at either position 238 or position 328 (by EU numbering).
  • 59. The antigen-binding molecule of claim 58, wherein the modified Fc region has Asp at position 238 or Glu at position 328.
  • 60. The antigen-binding molecule of claim 58, wherein at least one of the following positions (EU numbering) in the modified Fc region is occupied by the indicated amino acid: Asp at position 233;either Tip or Tyr at position 234;any one of Ala, Asp, Glu, Leu, Met, Phe, Tip, or Tyr at position 237;Asp at position 239;any one of Ala, Gln, or Val at position 267;any one of Asn, Asp, or Glu at position 268;Gly at position 271;any one of Ala, Asn, Asp, Gln, Glu, Leu, Met, Ser, or Thr at position 326;any one of Arg, Lys, or Met at position 330;any one of Ile, Leu, or Met at position 323;Asp at position 296.
  • 61. A pharmaceutical composition comprising the antigen-binding molecule of claim 35.
  • 62. A pharmaceutical composition comprising the antigen-binding molecule of claim 36.
  • 63. A method of reducing the concentration of an aggregated form of an antigen in a subject's plasma, the method comprising contacting the subject's plasma with an effective amount of the antigen-binding molecule of claim 35, wherein the antigen-binding molecule binds to the aggregated form of the antigen and facilitates removal of the aggregated form of the antigen from the subject's plasma.
  • 64. The method of claim 63, wherein the antigen-binding molecule preferentially clears the aggregated form of the antigen from the subject's plasma, as compared to the unaggregated form of the antigen.
  • 65. A method for producing an antigen-binding molecule, the method comprising: (a) culturing a cell comprising DNA encoding an antigen-binding molecule comprising an antigen-binding domain and an Fc region, wherein the antigen-binding domain binds to an aggregated form of an antigen with a binding strength that varies depending on pH, calcium ion concentration, or both pH and calcium ion concentration, and wherein the antigen naturally aggregates in plasma in vivo to produce the aggregated form; and(b) collecting the antigen-binding molecule from the cell culture.
  • 66. The method of claim 65, further comprising conducting an assay demonstrating that, at a pH between 6.7 and 10.0, the antigen-binding molecule binds more strongly to the aggregated form of the antigen than to an unaggregated form of the antigen.
  • 67. The method of claim 65, further comprising conducting an assay demonstrating that, at a pH between 6.7 and 10.0, a first complex comprising the antigen-binding molecule and the aggregated form of the antigen binds to a human Fcγ or FcRn receptor more strongly than does a second complex comprising the antigen-binding molecule and an unaggregated form of the antigen.
  • 68. A method for producing an antigen-binding molecule, the method comprising: (a) culturing a cell comprising DNA encoding an antigen-binding molecule comprising (i) an antigen-binding domain that binds to an aggregated form of an antigen, and (ii) an Fc region, thereby producing a cell culture comprising the antigen-binding molecule; and(b) collecting the antigen-binding molecule from the cell culture;
  • 69. The method of claim 68, further comprising conducting an assay demonstrating that, at a calcium concentration between 100 μM and 10 mM, the antigen-binding molecule binds more strongly to the aggregated form of the antigen than to an unaggregated form of the antigen.
  • 70. The method of claim 68, further comprising conducting an assay demonstrating that, at a calcium concentration between 100 μM and 10 mM, a first complex comprising the antigen-binding molecule and the aggregated form of the antigen binds to a human Fcγ or FcRn receptor more strongly than does a second complex comprising the antigen-binding molecule and an unaggregated form of the antigen.
  • 71. A method of screening for antigen-binding molecules that can remove an aggregated form of an antigen from plasma, the method comprising (a) providing an antigen-binding molecule that binds to the aggregated form of the antigen;(b) assaying binding of the antigen-binding molecule to the aggregated form of the antigen at a first pH between 6.7 and 10.0;(c) assaying binding of the antigen-binding molecule to the aggregated form of the antigen at a second pH between 4.0 and 6.5;(d) determining that the antigen-binding molecule binds more strongly to the aggregated form of the antigen at the first pH than at the second pH; and(e) selecting the antigen-binding molecule, based on the determination of (d).
  • 72. The method of claim 71, further comprising conducting at least one of the following assays: an assay to determine whether, at a pH between 6.7 and 10.0, the antigen binding molecule binds to the aggregated form of the antigen more strongly than to the unaggregated form of the antigen;an assay to determine whether, at a pH between 6.7 and 10.0, a first complex comprising the antigen-binding molecule and the aggregated form of the antigen binds to a human Fcγ or FcRn receptor more strongly than does a second complex comprising the antigen-binding molecule and an unaggregated form of the antigen;an assay to determine whether, at a calcium ion concentration between 100 μM and 10 mM, the antigen-binding molecule binds to the aggregated form of the antigen more strongly than to the unaggregated form of the antigen;an assay to determine whether, at a calcium ion concentration between 100 μM and 10 mM, a first complex comprising the antigen-binding molecule and the aggregated form of the antigen binds to a human Fcγ or FcRn receptor more strongly than does a second complex comprising the antigen-binding molecule and an unaggregated form of the antigen.
  • 73. A method of screening for antigen-binding molecules that can remove an aggregated form of an antigen from plasma, the method comprising (a) providing an antigen-binding molecule that binds to the aggregated form of the antigen;(b) assaying binding of the antigen-binding molecule to the aggregated form of the antigen at a first calcium ion concentration between 100 μM and 10 mM;(c) assaying binding of the antigen-binding molecules to the aggregated form of the antigen at a second calcium ion concentration between 0.1 μM and 30 μM;(d) determining that the antigen-binding molecule binds more strongly to the aggregated form of the antigen at the first calcium ion concentration than at the second calcium ion concentration; and(e) selecting the antigen-binding molecule, based on the determination of (d).
  • 74. The method of claim 73, further comprising conducting at least one of the following assays: an assay to determine whether, at a pH between 6.7 and 10.0, the antigen binding molecule binds to the aggregated form of the antigen more strongly than to the unaggregated form of the antigen;an assay to determine whether, at a pH between 6.7 and 10.0, a first complex comprising the antigen-binding molecule and the aggregated form of the antigen binds to a human Fcγ or FcRn receptor more strongly than does a second complex comprising the antigen-binding molecule and an unaggregated form of the antigen;an assay to determine whether, at a calcium ion concentration between 100 μM and 10 mM, the antigen binding molecule binds to the aggregated form of the antigen more strongly than to the unaggregated form of the antigen; oran assay to determine whether, at a calcium ion concentration between 100 μM and 10 mM, a first complex comprising the antigen-binding molecule and the aggregated form of the antigen binds to a human Fcγ or FcRn receptor more strongly than does a second complex comprising the antigen-binding molecule and an unaggregated form of the antigen.
Priority Claims (1)
Number Date Country Kind
2012-123782 May 2012 JP national
PCT Information
Filing Document Filing Date Country Kind
PCT/JP2013/064979 5/30/2013 WO 00