VARIANT DOMAIN ANTIBODIES

Abstract
The present invention relates to domain antibodies which have modified framework regions such that the potency or level of binding of the domain antibody is improved. In particular the present invention relates to domain antibodies which bind TNFa in which changes have been made to framework sequences and to constructs including these domain antibodies.
Description
FILING DATA

This application is associated with and claims priority from Australian patent application no. 2008904175 filed on 14 Aug. 2008, U.S. patent application No. 61/089,019 filed on 14 Aug. 2008 and Australian patent application no. 2009902142 filed on 13 May 2009, the entire contents of each of these applications are incorporated herein by reference.


FIELD

The present invention relates to domain antibodies which have modified framework regions. In particular the present invention relates to domain antibodies which bind TNFα and to constructs including these domain antibodies.


BACKGROUND

The smallest naturally-occurring functional binding component derived from an antibody is the single variable domain, also known as ‘domain antibody’ (or ‘dAb’). This may be the variable domain of the heavy or light chain of an antibody (termed ‘VH’ or ‘VL’). The functional ability of isolated variable domains was first demonstrated when a cloned repertoire of murine VH genes were expressed in bacteria and shown to interact specifically with one or more antigens (Ward et al., 1989).


Single variable-like domains occur naturally in camelids (known as VHH) and cartilaginous fish (e.g. V-NARs in sharks) mounted on Fc-equivalent constant domains (Hamers-Casterman et al., 1993, Dooley and Flajnik, 2005 and Streltsov et al., 2004) and are able to bind to cognate antigen without a complementary VL domain (Muyldermans et al., 1994). In humans, the natural occurrence of such single chain immunoglobulin molecules is linked to the unbalanced synthesis of heavy and light chains and is associated with malignant disorders, such as multiple myeloma (Jones, 1848).


Development of combinatorial libraries using variable gene chain shuffling and selection methods, such as phage display have made it possible to generate human dAb libraries from which human variable domains specific for a range of antigens have been selected. The resultant dAbs have avoided aggregation problems associated with isolating human domains that would normally be complementarily paired. Further, low immunogenicity afforded by the utilization of human variable domains has empowered the development of dAbs for therapeutic applications (Holt et al., 2003).


Domain antibodies represent good candidates for therapeutic agents as they can be engineered to specifically target molecules associated with disease. They have the additional benefits of being well expressed in bacteria, yeast and mammalian systems. Being of a small size (ranging from 11 kDa to 15 kDa), domain antibodies have potential for enhanced tissue penetration. Moreover, therapeutically important serum half lives have been engineered into domain antibody constructs (WO 04/058820A2).


Tumour necrosis factor alpha, or TNFα, is a multi-functional cytokine that plays a central role in immune regulation, infection and inflammation. While normal levels of TNFα are important to immune homeostasis, excess production of TNFα has been implicated in the pathogenesis of numerous diseases including rheumatoid arthritis, Crohn's disease and psoriasis. Blocking the activity of excess TNFα is established as an effective therapeutic strategy.


Domain antibodies targeted to human TNFα have been described. These include a human VL dAb (WO 2005/035572), VHH derived dAb (WO 2004/041862) as well as a TNFα dAb consisting of the framework of a VH domain used as a scaffold to display TNFα targeted peptides (Qin et al., 2007).


Engineering efforts aimed at improving antigen binding affinity of antibodies and dAbs are more commonly focused on hypervariable or complementarity determining regions (known as ‘CDRs’) that make specific contacts with the antigen. The CDRs form extended 0 loops that are exposed on the surface of the variable domain to provide a complementary surface for antigen binding. The three CDRs are flanked by four framework regions of conserved sequence that fold into two 13 sheets. The framework residues provide a scaffold for mounting the CDR loops and may also directly or indirectly influence antigen binding by positioning of the CDR loops (Fischmann et al., 1991 and Tulip et al., 1992). Some framework residues may directly alter the conformation of CDR loops by making atomic interactions between framework and CDR residues (Kettleborough et al., 1991, Foote and Winter, 1992 and Xiang et al., 1995).


This present invention is relevant in the field of antibody engineering. This invention particularly describes domain antibodies comprising specific framework variants that possess improved function. The invention is further relevant in the development of domain antibodies for targeting human TNFα and for the development of such domain antibodies for therapeutic applications.


Bibliographic details of the publications referred to by author in this specification are collected alphabetically at the end of the description.


Reference to any prior art in this specification is not, and should not be taken as, an acknowledgment or any form of suggestion that this prior art forms part of the common general knowledge in Australia.


SUMMARY OF THE INVENTION

In a first aspect the present invention provides a modified dAb in which the framework sequences of the unmodified dAb are as set out in SEQ ID NO 1 and wherein the modified dAb includes at least one substitution selected from the group consisting of D1A, D1Y, D1K, I2L, I2P, Q3Y, Q3F, Q3D, Q3H, S7A, S7G, S7D, S7Q, S7N, S9A, S10A, S10L, L11Q, S12A, S12L, S12V, S12T, S12Q, S14A, D17Y, D17H, R18D, R185, T20Y, T22A, T22Y, T22Q, T22H, T22K, Q38H, P40D, K42A, K425, K42H, P44L, K45S, K45L, K45Y, K45D, K45H, 148A, Y49S, Y49D, Y49H, G57A, G57S, G57L, G57Y, G57Q, G57H, G57K, P59A, P595, P59D, P59H, S60A, S60G, S60L, S60Y, S60D, S60Q, S60H, S60P, S60K, R61H, F62Y, T69D, D70Q, L79A, L79G, L795, L79Y, L79V, L79D, L79Q, L79H, L79K, F83A, F83S, F83L, F83Y, F83D and conservative substitutions thereof and combinations thereof.


In a second aspect the present invention provides a dAb which binds TNFα, the dAb having SEQ ID NO 1 wherein at least one amino acid in the framework regions is substituted, wherein the dAb has improved TNFα neutralizing potency in comparison to the dAb of SEQ ID NO 1.


In a third aspect the present invention provides a dAb which binds TNFα, the dAb having SEQ ID NO 1 wherein at least one amino acid in the framework regions is substituted, wherein the dAb has improved TNFα binding and/or improved protein expression levels in comparison to the dAb of SEQ ID NO 1.


In a fourth aspect the present invention provides a VL domain antibody comprising a VL framework acceptor sequence wherein the framework sequence has been modified such that the framework comprises at least one amino acid substitution at least one position selected from the group consisting of 3, 7, 12, 60, 79, 83 and combinations thereof.


In a fifth aspect the present invention provides a nucleic acid molecule encoding the dAb of the first, second or third aspects of the present invention.


In a sixth aspect the present invention provides a transformed cell comprising the nucleic acid molecule of the fifth aspect of the present invention.





BRIEF DESCRIPTION OF THE FIGURES


FIG. 1. TNFα binding ELISA data of Y49 variants. A representative histogram of TNFα binding ELISA showing resultant binding of variants with substitutions at position forty-nine.



FIG. 2. Representative TNFα neutralization data obtained using the high throughput 5-point format of the L929 neutralization assay.



FIG. 3. Ferritin binding ELISA data of the Ferritin dAb Fc variants. In the key, ‘Fe dAb Fc’ denotes Ferritin dAb Fc proteins.





DETAILED DESCRIPTION

The present invention relates to dAbs in which one or more substitutions have been made in the framework region of the dAb in order to improve antigen binding efficiency, neutralizing potency or other characteristics.


In a first aspect the present invention provides a modified dAb in which the framework sequences of the unmodified dAb are as set out in SEQ ID NO 1 and wherein the modified dAb includes at least one substitution selected from the group consisting of D1A, D1Y, D1K, I2L, I2P, Q3Y, Q3F, Q3D, Q3H, S7A, S7G, S7D, S7Q, S7N, S9A, S10A, S10L, L11Q, S12A, S12L, S12V, S12T, S12Q, S14A, D17Y, D17H, R18D, R185, T20Y, T22A, T22Y, T22Q, T22H, T22K, Q38H, P40D, K42A, K42S, K42H, P44L, K45S, K45L, K45Y, K45D, K45H, I48A, Y49S, Y49D, Y49H, G57A, G57S, G57L, G57Y, G57Q, G57H, G57K, P59A, P59S, P59D, P59H, S60A, S60G, S60L, S60Y, S60D, S60Q, S60H, S60P, S60K, R61H, F62Y, T69D, D70Q, L79A, L79G, L79S, L79Y, L79V, L79D, L79Q, L79H, L79K, F83A, F83S, F83L, F83Y, F83D and conservative substitutions thereof and combinations thereof.


In the three symbol code used, the first letter stands for the amino acid in the unmodified dAb construct, the last letter stands for the substitution amino acid and the number in the middle is the residue position. For example, S60A describes a dAb variant with single substitution at position 60, where the Serine in the unmodified dAb is replaced by an Alanine. References herein to amino acid positions of unmodified dAb refer to the numbering of amino acids as defined in the Kabat database of Sequences of Proteins of Immunological Interest (“Sequences of Proteins of Immunological Interest”; US Department of Health and Human Services). The Kabat numbering is typically referred to in the art as the ‘Kabat convention’. Variants described in this invention that comprise of multiple amino acid substitutions are denoted by a series of the three symbol code separated by ‘_’. For example, S60A_F83Y is a dAb variant comprising double substitutions where the Serine and Phenylalanine that were found in positions 60 and 83 of the unmodified dAb were replaced by an Alanine and a Tyrosine, respectively.


In the sequence set out in SEQ ID NO 1 the framework regions are residues 1-23, 35-49, 57-88 and 98-108.


In various forms of the invention the modified domain antibody comprises a substitution selected from the group consisting of L79G, L79S, L79Y, L79K, L79D, L79A, L79Q, L79V and L79H; and/or a substitution selected from the group consisting of S60G, S60A, S60D, S60L, S60H, S60Q, S60Y, S60P and S60K; and/or a substitution selected from the group consisting of S12T, S12L, S12V, S12A and S12Q; and/or a substitution selected from the group consisting of F83Y, F83L, F83A, F83S and F83D; and/or a substitution selected from the group consisting of P59A, P59S, P59D and P59H; and/or a substitution selected from the group consisting of G57S, G57Q, G57H, G57A, G57L, G57Y and G57K; and/or a substitution selected from the group consisting of Q3F, Q3Y, Q3H and Q3D; and/or a substitution selected from the group consisting of S7A, S7N, S7G, S7Q and S7D.


In a second aspect the present invention provides a dAb which binds TNFα, the dAb having SEQ ID NO 1 wherein at least one amino acid in the framework regions is substituted, wherein the dAb has improved TNFα neutralizing potency in comparison to the dAb of SEQ ID NO 1.


In a preferred embodiment of the second aspect of the invention the substitution is selected from the group consisting of D1A, D1Y, D1K, I2L, I2P, Q3F, Q3Y, Q3D, Q3H, S7A, S7D, S7Q, S7G, S7N, S9A, S10A, S10L, S12A, S12L, S12Q, S12V, S12T, S14A, R18D, T22A, K42A, K45S, Y49H, G57S, G57Q, G57H, P59A, P59S, P59D, S60G, S60A, S60L, S60D, S60Q, S60H, F62Y, L79G, L79S, L79Y, L79D, L79K, F83A, F83L, F83S, F83Y and conservative substitutions thereof and combinations thereof.


In various forms of the invention the domain antibody comprises a substitution selected from the group consisting of L79G, L79S, L79Y, L79K and L79D; and/or a substitution selected from the group consisting of S60G, S60A, S60D, S60L, S60H and S60Q; and/or a substitution selected from the group consisting of S12T, S12L, S12V, S12A and S12Q; and/or a substitution selected from the group consisting of F83Y, F83L, F83A and F83S; and/or a substitution selected from the group consisting of P59A, P59S and P59D; and/or a substitution selected from the group consisting of G57S, G57Q and G57H; and/or a substitution selected from the group consisting of Q3F, Q3Y, Q3H and Q3D; and/or a substitution selected from the group consisting of S7A, S7N, S7G, S7Q and S7D.


The domain antibody may include multiple substitutions. These multiple substitutions may be selected from the group consisting of S60A_F83Y, Q3Y_S12L, S12L_S60A_L79Y_F83Y, S7Q_S60A, S7Q_S12L, Q3Y_S7Q, S12L_L79Y_F83Y, Q3Y_S60A, S12L_S60A_F83Y, S7Q_L79Y_F83Y, Q3Y_S12L_S60A_F83Y, Q3Y_S7Q_S12L_S60A, Q3Y_S60A_F83Y, Q3Y_S7Q_S12L, Q3Y_S7Q_S12L_L79Y, Q3Y_S12L_L79Y, Q3Y_L79Y, S7Q_S12L_S60A_F83Y, L79Y_F83Y, Q3Y_S12L_L79Y_F83Y, S60A_L79Y_F83Y, S7Q_S12L_L79Y_F83Y, Q3Y_S7Q_S12L_S60A_L79Y_F83Y, Q3Y_S7Q_S60A, Q3Y_S7Q_S60A_L79Y and S12L_S60A_L79Y.


In a third aspect the present invention provides a dAb which binds TNFα, the dAb having SEQ ID NO 1 wherein at least one amino acid in the framework regions is substituted, wherein the dAb has improved TNFα binding and/or improved protein expression levels in comparison to the domain antibody of SEQ ID NO 1.


In a preferred embodiment of the third aspect of the invention the substitution is selected from the group consisting of S7A, L11Q, D17Y, D17H, R185, T20Y, T22Y, T22Q, T22H, T22K, Q38H, P40D, K42S, K42H, P44L, K45L, K45Y, K45D, K45H, 148A, Y49S, Y49D, Y49H, G57A, G57S, G57L, G57Y, G57Q, G57H, G57K, P59S, P59D, P59H, S60A, S60L, S60Y, S60D, S60Q, S60H, S60P, S60K, R61H, T69D, D70Q, L79A, L79S, L79Y, L79D, L79Q, L79H, L79K, F83A, F83Y, F83D and combinations thereof.


It is preferred that the substitution is selected from the group consisting of S7A, Y49H, G57S, G57Q, G57H, P59S, P59D, S60A, S60L, S60D, S60Q, S60H, L795, L79Y, L79D, L79K, F83A, F83Y and combinations thereof.


In a fourth aspect the present invention provides a VL domain antibody comprising a VL framework acceptor sequence wherein the framework sequence has been modified such that the framework comprises at least one amino acid substitution at least one position selected from the group consisting of 3, 7, 12, 60, 79, 83 and combinations thereof.


In a preferred embodiment of this aspect of the present invention the domain antibody comprises at least one framework substitution selected from the group Y at position 3, Q at position 7, L at position 12, A at position 60, Y at position 79, Y at position 83 and combinations thereof.


As mentioned above the reference position number in the fourth aspect uses the Kabat numbering convention. As will be well known to persons skilled in this art a number databases providing VL and VH framework acceptor sequences are readily available. Examples of these databases include the Kabat (www.kabatdatabase.com/; Johnson and Wu, 2001), IMGT (http://imgt.cines.fr/; Lefranc, 2003) and VBase (http://vbase.mrc-cpe.cam.ac.uk/; Retter et al., 2005) databases.


In a preferred embodiment, the dAb is attached to an Fc region or a modified Fc region such as described in WO 2007/087673, (the disclosure of which is incorporated by reference) with or without the cysteine substitution. The sequence of the unmodified anti-TNFα dAb attached to the modified Fc is set out in SEQ ID NO 2. It is further preferred that the truncated CH1 and modified hinge is XEPKSZDKTHTCPPCPA (SEQ ID No: 3) wherein X is valine, leucine or isoleucine and Z is absent or an amino acid other than cysteine. In certain embodiments the modified Fc is SEQ ID NO 6 or SEQ ID NO 7.


When the dAb is attached to other domains, then the protein in its entirety is herein referred to as a ‘dAb construct’.


In a fifth aspect the present invention provides a nucleic acid molecule encoding the dAb of the first, second or third aspects of the present invention.


In a sixth aspect the present invention provides a transformed cell comprising the nucleic acid molecule of the fourth aspect of the present invention wherein the cell comprises the nucleic acid molecule.


The transformed cell may be either a prokaryotic or eukaryotic cell. Where the cell is a bacterial cell the bacteria itself may be used therapeutically as described in US 2005/0101005 and WO 2000/023471, the disclosures of which are incorporated herein by reference.


In certain embodiments the nucleic acid molecule has a sequence as set out in SEQ ID NO 8 to SEQ ID NO 708.


The present invention describes improved dAbs in which substitutions have been made in the framework regions. This highlights the importance particular framework positions play in improving potency and other characteristics. The dAbs of the present invention display improved function when substitutions at particular positions are made with amino acids of more than one amino acid class. This invention teaches the value of strong consideration of amino acid utilized at the positions of 1, 3, 7, 12, 57, 59, 60, 79 and 83 in a variable immunoglobulin domain (using the Kabat numbering convention). These positions are herein referred to as ‘robust’.


In yet another aspect, improved dAbs are variants at positions 9, 10, 12, 14, 18, 42, 79 and 83 in VL dAbs. These are dAb variants where the described framework substitutions occur at a residues whose atoms lie more than six Ångstroms (6 Å) from those CDR residues. Prior art teaches that these residues being spatially distant from CDR residues are unlikely to interact directly with the antigen, and thus unlikely to affect the interaction between the dAb and its antigen.


As described herein, a “domain antibody” or “dAb” refers to a single immunoglobulin variable domain that specifically binds a given antigen. A dAb binds antigen independently of any other domains; however, as the term is used, a dAb can be present in a homo- or heteromultimer with other domains where the other domains are not required for antigen binding by the dAb, i.e. where the dAb binds antigen independently of the additional domains. The dAb may be a heavy chain variable domain (VH) or a light chain variable domain (VL). A dAb describes the one single variable domain that is one of many domains found in an antibody molecule. The dAb of this present invention is a VL, further it is of kappa subclass (Vκ).


A domain antibody or a domain antibody construct is not an antibody. A domain antibody or dAb is one domain within an antibody. An “antibody” is a tetrameric multi-domain glycoprotein. An antibody consists of two identical pairs of immunoglobulin chains, each pair having one light and one heavy chain. The light immunoglobulin chain, which can be of either kappa or lambda subclass (Vκ or Vλ), comprises two domains, a N-terminal variable domain (VL) and one constant domain at the C-terminus (CL), while the heavy chain contains four modular domains, a N-terminal variable domain (VH) followed by three different constant domains (CH1, CH2 and CH3). The variable domains of the light and heavy chains are together responsible for binding to an antigen. The antigen binding surface of an antibody is afforded by the two variable domains, whereas that of a dAb is present within the one single domain of the dAb. It is within the scope of this present invention that the improved dAb described herein may be relevant to a subset of antibody molecules whose antigen binding surface is comprised entirely of only one of the two variable domains.


A domain antibody, whether it is in isolation, within a dAb construct or an antibody, consists of four “framework” regions separated by three hypervariable or complementarity-determining regions (or CDRs). The extents of the framework regions and CDRs in variable domains have been defined (see Kabat et al., “Sequence of Proteins of Immunological Interest”, US Department of Health and Human Services). The sequences of the framework regions of different light and heavy chains are relatively conserved within a species. Framework residues form the scaffold that positions and supports the CDRs that in turn dictate the antigen specificity and binding capacity. The CDRs are primarily responsible for antigen binding. The CDRs of each heavy and light chain are typically referred to as CDR1, CDR2 and CDR3, numbered sequentially starting from the N-terminus and are also typically identified by the chain in which the particular CDR is located. Thus, a VL CDR1 is the CDR1 from the variable domain of the light chain.


As described in this invention, “framework variants” refers herein to variable domains or dAbs which differ in amino acid sequence from an unmodified variable domain or dAb by virtue of substitution of one or more amino acid residue(s) in the unmodified sequence. The present invention describes framework variants. The variants are within the framework regions regardless of how the framework regions and CDRs are defined. For example: whether the definition utilizes the sequence-based alignment of the Kabat convention, or the structural-based definition of Chothia, or the IMGT and AHo schemes that unify the alignment of antibody immunoglobulin domains with T-cell receptor domains (Kabat et al., 1983, Chothia and Lesk, 1987, Lefranc et al., 2003 and Honegger and Pluckthun, 2001). The amino acid residue numbering used to describe the present invention follows that defined by the Kabat convention.


As used herein, a “protein” and “polypeptide” are used interchangeably and include reference to a chain of amino acid residues.


The amino acids referred herein are described: Alanine (A), Arginine (R), Asparagine (N), Aspartic Acid (D), Cysteine (C), Glutamic Acid (E), Glutamine (Q), Glycine (G), Histidine (H), Isoleucine (I), Leucine (L), Lysine (K), Methionine (M), Phenylalanine (F), Proline (P), Serine (S), Threonine (T), Tryptophan (W), Tyrosine (Y) and Valine (V).


Amino acid “substitution” refers to the replacement of the original amino acid in the unmodified protein sequence by a second amino acid at the same position in the protein sequence. The framework variants of this invention are substitution variants. That is, the specific residue or amino acid, of the unmodified variable domain or dAb is replaced by one of the other nineteen naturally occurring amino acids. This substitution does not alter the length of the polypeptide or protein sequence. The variants described in this invention are not insertion or deletion variants where the length of the polypeptide may be altered.


A substitution is deemed to be “conservative” when the original amino acid and the second or replacement amino acid have similar chemical and/or physical properties (e.g., charge, structure, polarity, hydrophobicity, hydrophilicity). As such, conservative substitutions do not substantially alter the activity of the protein. Conservative substitutions providing functionally similar amino acids are well known in the art. The following classes each contain amino acids that are conservative substitutions for one another:


(1) A, G and S


(2) N, Q, S, T, Y, K, R, H, D and E


(3) Q, N


(4) D, E


(5) K, R, H


(6) A, V, L, I, P, F, W, M, C, G


(7) Y, F, W


(8) C, S and T.


A “robust” residue or position within a protein referred herein are those residues or positions that can be substituted into amino acids of more than one amino acid class as defined above as conservative amino acids and still retain function.


The term “hotspot” means a portion of the protein sequence of a CDR or of a framework region of a variable domain which is a site of increased variation. Although CDRs are themselves considered to be regions of hypervariability, it has been learned in the present invention that particular sites, or hotspots, can be located in the framework regions which undergo certain substitution mutations.


The improved dAbs of this invention are of an immunoglobulin variable light chain domain or VL. It is of the Vκ subclass. As the invention relates to substitution of amino acids in the framework regions of the dAb and it is the CDRs, not framework residues that comprise the antigen binding surface, the dAbs described in the invention may be broadly applied to VL dAbs of all antigen specificity. The antigen may be human or non-human, protein, nucleic acid, lipid or carbohydrate.


The improved dAbs of this invention are of a VL domain of the Vκ subclass. Immunoglobulin variable light chain domains may be of either Vκ or Vλ, subclass. It is within the scope of the present invention that the improved dAbs may be also applicable in a VL dAb of lambda subclass.


The improved dAbs provided in this present invention are human dAbs. That is, the sequence of the unmodified dAb is one of a human light chain variable domain. The improved dAbs may also be applied to a humanized dAb, a CDR-grafted dAb, a synhumanised dAb, a primatized dAb, a camelid derived antibody fragment and variants thereof.


The improved dAb variants of this present invention may be utilized in isolation or be attached to other domains, e.g. comprising only a part of the protein. In a preferred embodiment of the present invention, the improved dAb is attached to a human or non-human primate heavy chain immunoglobulin constant region. The human or non-human primate heavy chain constant region may be selected from a group consisting of IgG1, IgG2, IgG3, IgG4, IgM, IgE and IgA (as well as subtypes thereof).


The improved dAb may be attached to other domains. The other attached domains may be antibody-based or antibody-like domains, such as another dAb, Fab fragments, scFv domains, VHH or V-NAR domains derived from camels, sharks and some fishes. The other domains may be non-antibody protein domains with a natural or artificially binding specificity. Naturally occurring protein domains with natural binding specificities that may be attached to the improved dAb include extracellular domains of receptors, such as those of the TNF receptor superfamily e.g. TNFRI (p55), TNFRII (p75), 95, DCR1, DCR2, DR3, DR4, DR5, DR6, EDAR, NGFR, RANK, LTβR, FN14, HVEM, CD27, CD30, CD40, 4-1BB, OX40, GITR, BCMA, TACI, BAFFR, XEDAR, TROY, RELT and soluble receptors such as osteoprotegerin and DCR3. Further others include growth hormones, glucagon-like peptide (GLP-1), interleukins (IL-2, -4, -5, -6, -7, -10, -12, -13, -14, -15, -16, -18, -21, -23, -31), FGF-basic, IGF-1, keratinocyte growth factor, insulin, LIF, GM-CSF, G-CSF, M-CSF, Epo and TPO, lymphotoxin, TRAIL, TGF-β, VEGF-2, leptin, interferons (IFN-α, -β and -γ), parathyroid hormone, Dornase alfa, TACE, thrombin, PDZ modules of signalling proteins, adhesion molecules and enzymes. Protein domains with artificially derived binding specificities include scaffolds based on transferrin (Transbody; WO 08/072,075A2), three-helix bundle from Z-domain of Protein A (Affibody®; WO 00/63243A1), human C-type lectin domain (Tetranectin; WO 98/56906A1), tenth fibronectin type III domain (AdNectin™; U.S. Pat. No. 6,818,418), Kunitz-type domain of trypsin inhibitor (WO 96/04378A2), ankyrin repeat protein, lipocalins (Anticalin®; WO 99/16873A1), 7-crystallin or ubiquitin molecule (Affilin™; WO 06/040129A2), trypsin inhibitor II (Microbody) and immunoglobulin-like domain derived from cytotoxic T lymphocyte-associated antigen 4 (CTLA-4), or modified variants thereof (Evibody™; U.S. Pat. No. 7,166,697). Further, attaching more than one domain together in a polypeptide can be achieved using linker polypeptides comprise two or more amino acid residues. Such linker polypeptides are well known in the art (see Holliger et al., 1993).


In another aspect, the improved dAbs may be conjugated to a protein such as serum albumin or a polymer (e.g. PEG). Alternatively, the dAbs may be immunoconjugated in which there is a covalent linkage of a therapeutic agent and can include cytotoxins such as native or modified Pseudomonas or Diphtheria toxin, calicheamicin and the like, encapsulating agents, (e.g. liposomes) which themselves contain pharmacological compositions, radioactive agents and other labels (e.g. enzyme reporter).


The invention relates to the specific amino acid substitution in a dAb that endows the dAb construct with improved function. The methods used to produce these mutations are wide and varied. The method utilized in the present embodiment is one of many routes that can be taken. In the case whereby the substitution(s) encompassed by the improved dAbs of this present invention are to be directly incorporated into a given human variable domain or domain antibody. The substitution may be introduced directly using standard techniques of molecular biology to prepare such DNA sequences. Substitution by site-directed mutagenesis and polymerase chain reaction (PCR) techniques is well known in the art. For example oligonucleotide directed synthesis such as that using a pre-existing variable region may be used (Jones et al., 1986, Verhoeyen et al., 1988 and Riechmann et al., 1988). Enzymatic filling of gapped nucleotide using T4 DNA polymerase may be employed (Queen et al., 1989 and WO 90/07861). This methodology will generate a small number of variants. To further increase the number of variants that can be produced at one time, one may utilize error-prone PCR to create a small library of substitution variants (Cadwell and Joyce, 1992, Hawkins et al., 1992, Fromant et al., 1995). Methods for generating high diversity libraries include DNA shuffling where recombination can be performed between variable genes or alternatively between variable gene libraries


(Stemmer et al., 1994, Harayama, 1998, Zha et al., 2003). Chain shuffling is an alternative method which involves the sequential replacement of the variable heavy and light chains (Kang et al., 1991b and Marks, 2004). In the case where large libraries of dAbs are employed for selection against a given target, natural libraries that use rearranged variable genes can be harvested from human B cells (Marks et al., 1991 and Vaughan et al., 1996) or synthetic libraries prepared from oligonucleotide cloning human immunoglobulin variable genes (Hoogenboom and Winter 1992, Barbas et al., 1992, Nissim et al., 1994, Griffiths et al., 1994 and de Kruif et al., 1995) including semi-synthetic libraries (Knappick et al., 2000).


The improved dAbs of the present invention may be identified by affinity maturation techniques. It is expected that those of skill in the art are knowledgeable in the numerous molecular engineering approaches and methods used to affinity mature antibodies and antibody-based fragments such as dAbs, Fabs, scFv and variations thereof. Typically, the CDRs are targeted in random mutagenesis (Jackson et al., 1995, van den Beucken et al., 2003, Zahnd et al., 2004, Lee et al., 2004, Yau et al., 2005) or targeted mutagenesis (Yelton et al., 1995, Schier et al., 1996, Thompson et al., 1996, Boder et al., 2005, Ho et al., 2005, Yoon et al., 2006). An example of targeted mutagenesis methodology is CDR walking involving a two-step process in which individual CDRs of the light and heavy chain are sequentially modified and the best candidates subsequently combined (Yang et al., 1995, Wu et al., 1998). Alternatively, mutations can be made across the entire sequence, for example to identify CDR and framework hotspots. This can be achieved by in vitro DNA amplification such as error-prone PCR (van den Beucken et al., 2003, Zhang et al., 2005, Thom et al., 2006, Finlay et al., 2009) or in vivo propagation of antibody genes within mutator bacterial strains (Irving et al., 1996 and Low et al., 1996, Coia et al., 2001). Targeting specific or hotspot residues for improvement can be determined experimentally using such techniques as alanine scanning (Cunnigham and Wells, 1989, Ashkenazi et al., 1990, Chatellier et al., 1995, Weiss et al., 2000, Koide et al., 2007) or homolog shotgun scanning (Vajdos et al., 2002, Murase et al., 2003, Pal et al., 2005) where the role of side chain functional groups at specific positions are identified.


When the three-dimensional structure of an antibody: antigen complex is available (e.g. obtained by x-ray crystallography), the molecular interactions between amino acids can facilitate the selection of amino acid residues for mutation. It is known that amino acid substitutions can change the three-dimensional structure of an antibody to improve function (Kast and Hilvert, 1997, Chen et al., 1999, Valjakka et al., 2002, Barderas et al., 2008). Three-dimensional models of antibodies and fragments thereof, such as domain antibodies described in this present invention, are commonly available and are familiar to those skilled in the art. Computer programs are available which illustrate and display probable three-dimensional conformational structures of selected candidate sequences. Inspection of these displays permits analysis of the likely role residues play in the recognition of the target antigen.


The improved dAbs may also be produced and selected against the target antigen using display techniques that are known to those skilled in the art. For example, bacteriophage lambda expression systems may be screened directly as bacteriophage plaques or as colonies of lysogens (Huse et al., 1989, Caton and Koprowski, 1990, Mullinax et al., 1990, Persson et al., 1991), or more commonly utilized, are selection display systems that enable a nucleic acid to be linked to the polypeptide it expresses. One of these methods is phage display techniques in which diverse protein sequences are displayed on the surface of filamentous bacteriophage, typically as fusions to bacteriophage coat proteins, or displayed externally on lambda phage capsids (phagebodies). Methods to undertake these processes are well known in the art (Scott and Smith, 1990, McCafferty et al., 1991, WO 92/01047, Kang et al., 1991a, Clackson et al., 1991, Lowman et al., 1991, Burton et al., 1991, Hoogenboom et al., 1991, Chang et al., 1991, Breitling et al., 1991, Marks et al., 1991, Barbas et al., 1992, Hawkins and Winter 1992, Marks et al., 1992, Lerner et al., 1992). Other systems for generating libraries of polypeptides involve the use of cell-free enzymatic machinery for the in vitro synthesis of the library members. For example, RNA display (Tuerk and Gold 1990, Ellington and Szostak, 1990), in vitro translation in stabilized polysome complexes (WO88/08453, WO90/05785), or in microcapsules formed by water-in-oil emulsions (WO99/02671, Tawfik and Griffiths, 1998).


The improved dAbs of this present invention may be characterized in a number of ways. One skilled in the art will appreciate the plethora of methods available. Some examples include, but are not limited to, attaching the antigen to a solid support (e.g. using an enzyme-linked immunoassay or by surface plasmon resonance analysis), in solution utilizing a labelling agent (e.g biotinylation, radiolabelling or fluorescent tagging) and employing depletion or subtraction procedures (e.g. FACS analysis; Daugherty et al., 1998, Ridgway et al., 1999, van den Beucken et al., 2003).


In another aspect, an assay for the activity or function of the target antigen may be assayed or quantified as a measurable activity or function of the target antigen. An assay for the activity or function of a target antigen includes for example, binding activity, cell activation, cell killing, cell proliferation, cell signalling, enzymatic activity, ligand-dependent internalization, promotion of cell survival and gene expression. The present invention describes framework variants that target the antigen TNFα; activity can be assayed using the TNFα-mediated cytotoxicity of L929 cells.


The term “neutralization” when used in reference to a molecule, means that the molecule interferes with a measurable activity or function of the target antigen. A molecule is neutralizing if it reduces a measurable activity or function of the target antigen. In several aspects of the present invention, where the target antigen is TNFα, neutralizing activity can be assessed using the L929 cytotoxicity assay. For example, murine L929 cells are susceptible to the cytotoxic effects of human TNFα in a dose-dependent manner. The ability to inhibit or block TNFα can be quantitated by the neutralization of the TNFα-mediated cytotoxicity of L929 cells. This ability to inhibit TNFα, or the activity as a TNFα inhibitor, is referred to as “potency”, such that the activity or potency of the improved dAbs of this invention refers to its ability to inhibit effects of TNFα in this assay.


Binding activity of the dAbs of this current invention for target antigen may be measured in a number of different assays. Such binding assays are well-known to those skilled in the art. These include enzyme-linked immunosorbent assay (ELISA) and analysis by surface plasmon resonance (SPR). Binding assays enable the measurement of the strength of the interaction between the two said molecules.


The term “dissociation constant” or “KD” refers to the strength of the interaction between two molecules, and in this invention, the affinity of dAb or dAb variant for its target antigen. The “KD” is the dissociation constant at equilibrium, and has units of Molarity. The affinity of an antibody or dAb for an antigen can be determined experimentally using any suitable method (eg. Berzofsky et al., 1984 and Kuby, 1992; and methods described herein).


As used herein “nucleic acid” or “nucleic acid molecule” includes reference to a deoxyribonucleotide or ribonucleotide polymer in either single-, double-, triple-stranded or any combination thereof. Nucleic acid molecules of the present invention can be in the form of RNA, such as mRNA, hnRNA, tRNA or any other form, or in the form of DNA, including, but not limited to, cDNA and genomic DNA obtained by cloning or produced synthetically, or any combinations thereof. Nucleic acid molecules of the present invention can include nucleic acid molecules comprising the coding sequence for a dAb, or dAb variant or specified portion; and nucleic acid molecules which comprise a nucleotide sequence different from those described above but which, due to the degeneracy of the genetic code, still encode at least one dAb or dAb variant as described herein and/or as known in the art. The genetic code is well known in the art. Thus, it would be routine for one skilled in the art to generate such degenerate nucleic acid variants that code for specific dAb or dAb variants or specified portion of the present invention.


By “transformed cell” is meant a cell that comprises the nucleic acid molecule of this present invention. Transformed cells may be prokaryotic cells such as E. coli or eukaryotic cells such as yeast or mammalian cells. The transformed cells themselves may be used therapeutically as described in US 2005/0101005 and WO 2000/023471, the disclosures of which are incorporated herein by reference.


Further, the transformed cell may be utilized in the production of the dAb or dAb variant Fc protein of the present invention. In such case, a suitable host cell system may be used for expression of the DNA sequences encoding for the improved dAbs. These may be bacteria, plant, yeast, insect and mammalian cells. It is expected that those of skill in the art are knowledgeable in the numerous expression systems for the expression of proteins including E. coli and other bacterial hosts and suitable mammalian host cells including COS-1 (e.g. ATCC CRL 1650), COS-7 (e.g. ATCC CRL-1651), HEK293, BHK21 (e.g. ATCC CRL-10), CHO (e.g. ATCC CRL 1610). BSC-1 (e.g. ATCC CRL-26) SP2/0-Ag14 (e.g. ATCC CRL 1851), HepG2 cells, YB2/0 cells, NS0, Per.C6, EBx, HeLa cells and the like, which are readily available from, for example, American Type Culture Collection (ATCC; Manassas, Va. Usa.)


Alternatively, nucleic acids of the present invention can be expressed in a host cell by turning on (by manipulation) in a host cell that contains endogenous DNA encoding an domain antibody or specified portion or variant of the present invention. Such methods are well known in the art, e.g., as described in U.S. Pat. Nos. 5,580,734, 5,641,670, 5,733,746, and 5,733,761, entirely incorporated herein by reference.


The present invention also provides a composition comprising the dAb of the present invention together with a pharmaceutically acceptable carrier.


The present invention also provides a method of treating a disorder characterized by human TNFα activity in a human subject comprising administering to the subject an effective amount of the dAb of the present invention or a composition containing such a dAb or dAb construct.


Typically the disorder characterized by human TNFα activity is selected from the group consisting of inflammation; inflammatory diseases; sepsis, including septic shock, endotoxic shock, gram negative sepsis and toxic shock syndrome; autoimmune disease, including rheumatoid arthritis, juvenile arthritis, rheumatoid spondylitis, ankylosing spondylitis, Sjögren's syndrome, osteoarthritis and gouty arthritis, allergy, multiple sclerosis, autoimmune diabetes, autoimmune uveitis, psoriasis, pemphigoid and nephrotic syndrome; inflammatory conditions of the eye, including macular degeneration, angiogenesis-related ocular disorder (in particular age-related macular degeneration), uveitis, Behçet's disease; infectious disease, including fever and myalgias due to infection and cachexia secondary to infection; graft versus host disease; tumour growth or metastasis, hematologic malignancies; pulmonary disorders including asthma, adult respiratory distress syndrome, shock lung, chronic pulmonary inflammatory disease, pulmonary sarcoidosis, pulmonary fibrosis and silicosis; inflammatory bowel disorders including Crohn's disease and ulcerative colitis; cardiac disorders, congestive heart failure; vascular disorders including Wegener's disease, giant cell arteritis; inflammatory bone disorders, central nervous system disorders such as Alzheimer's disease; peripheral nervous system disorders such as sciatica, hepatitis, coagulation disturbances, burns, reperfusion injury, endometriosis, keloid formation and scar tissue formation.


The route of administration may be intravenous, intramuscular, bolus, intraperitoneal, subcutaneous, respiratory, inhalation, topical, nasal, vaginal, rectal, buccal, sublingual, intranasal, subdermal, and transdermal.


Typically, treatment of pathologic conditions is effected by administering an effective amount or dosage of at least one dAb composition that total, on average, a range from at least about 0.01 to 500 milligrams of at least one dAb, specified portion or variant/kilogram of patient per dose, and, preferably, from at least about 0.1 to 100 milligrams of dAb, specified portion or variant/kilogram of patient per single or multiple administration, depending upon the specific activity of dAb, specified portion or variant contained in the composition. Alternatively, the effective serum concentration can comprise 0.1-5000 μg/ml serum concentration per single or multiple administrations. Suitable dosages are known to medical practitioners and will, of course, depend upon the particular disease state, specific activity of the composition being administered, and the particular patient undergoing treatment. In some instances, to achieve the desired therapeutic amount, it can be necessary to provide for repeated administration, i.e., repeated individual administrations of a particular monitored or metered dose, where the individual administrations are repeated until the desired daily dose or effect is achieved. Preferred doses can optionally include 0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99 and/or 100 mg/kg/administration, or any range, value or fraction thereof, or to achieve a serum concentration of 0.1, 0.5, 0.9, 1.0, 1.1, 1.2, 1.5, 1.9, 2.0, 2.5, 2.9, 3.0, 3.5, 3.9, 4.0, 4.5, 4.9, 5.0, 5.5, 5.9, 6.0, 6.5, 6.9, 7.0, 7.5, 7.9, 8.0, 8.5, 8.9, 9.0, 9.5, 9.9, 10, 10.5, 10.9, 11, 11.5, 11.9, 20, 12.5, 12.9, 13.0, 13.5, 13.9, 14.0, 14.5, 4.9, 5.0, 5.5, 5.9, 6.0, 6.5, 6.9, 7.0, 7.5, 7.9, 8.0, 8.5, 8.9, 9.0, 9.5, 9.9, 10, 10.5, 10.9, 11, 11.5, 11.9, 12, 12.5, 12.9, 13.0, 13.5, 13.9, 14, 14.5, 15, 15.5, 15.9, 16, 16.5, 16.9, 17, 17.5, 17.9, 18, 18.5, 18.9, 19, 19.5, 19.9, 20, 20.5, 20.9, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 96, 100, 200, 300, 400, 500, 600, 700, 800, 900, 1000, 1500, 2000, 2500, 3000, 3500, 4000, 4500, and/or 5000 μg/ml serum concentration per single or multiple administration, or any range, value or fraction thereof.


Alternatively, the dosage administered can vary depending upon known factors, such as the pharmacodynamic characteristics of the particular agent, and its mode and route of administration; age, health, and weight of the recipient; nature and extent of symptoms, kind of concurrent treatment, frequency of treatment, and the effect desired. Usually, a dosage of active ingredient can be about 0.1 to 100 mg/kg of body weight. Ordinarily, 0.1 to 50, and, preferably, 0.1 to 10 mg/kg per administration or in sustained release form is effective to obtain desired results.


As a non-limiting example, treatment of humans or animals can be provided as a one-time or periodic dosage of at least one dAb, specified portion or variant of the present invention, 0.1 to 100 mg/kg, such as 0.5, 0.9, 1.0, 1.1, 1.5, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 40, 45, 50, 60, 70, 80, 90 or 100 mg/kg, per day, on at least one of day 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, or 40, or, alternatively or additionally, at least one of week 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, or 52, or, alternatively or additionally, at least one of 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or years, or any combination thereof, using single, infusion or repeated doses.


Dosage forms (composition) suitable for internal administration generally contain from about 0.1 milligram to about 500 milligrams of active ingredient per unit or container. In these pharmaceutical compositions, the active ingredient will ordinarily be present in an amount of about 0.5-99.999% by weight based on the total weight of the composition.


For parenteral administration, the dAb, specified portion or variant can be formulated as a solution, suspension, emulsion or lyophilized powder in association, or separately provided, with a pharmaceutically acceptable parenteral vehicle. Examples of such vehicles are water, saline, Ringer's solution, dextrose solution, and 1-10% human serum albumin. Liposomes and nonaqueous vehicles, such as fixed oils, may also be used. The vehicle or lyophilised powder may contain additives that maintain isotonicity (e.g., sodium chloride, mannitol) and chemical stability (e.g., buffers and preservatives). The formulation is sterilized by known or suitable techniques.


Suitable pharmaceutical carriers are described in the most recent edition of Remington's Pharmaceutical Sciences, A. Osol, a standard reference text in this field.


Domain antibodies of the present invention can be delivered in a carrier, as a solution, emulsion, colloid, or suspension, or as a dry powder, using any of a variety of devices and methods suitable for administration by inhalation or other modes described herein or known in the art.


Formulations for parenteral administration can contain as common excipients sterile water or saline, polyalkylene glycols, such as polyethylene glycol, oils of vegetable origin, hydrogenated naphthalenes and the like. Aqueous or oily suspensions for injection can be prepared by using an appropriate emulsifier or humidifier and a suspending agent, according to known methods. Agents for injection can be a non-toxic, non-orally administrable diluting agent, such as aqueous solution or a sterile injectable solution or suspension in a solvent. As the usable vehicle or solvent, water, Ringer's solution, isotonic saline, etc. are allowed; as an ordinary solvent, or suspending solvent, sterile involatile oil can be used. For these purposes, any kind of involatile oil and fatty acid can be used, including natural, synthetic or semisynthetic fatty oils or fatty acids; natural, synthetic or semisynthetic mono- or di- or tri-glycerides. Parenteral administration is known in the art and includes, but is not limited to, conventional means of injections, a gas pressured needle-less injection device as described in U.S. Pat. No. 5,851,198, and a laser perforator device as described in U.S. Pat. No. 5,839,446, entirely incorporated herein by reference.


The invention further relates to the administration of at least one dAb, specified portion or variant by parenteral, topical, subcutaneous, intramuscular, intravenous, intraarticular, intrabronchial, intraabdominal, intracapsular, intracartilaginous, intracavitary, intracelial, intracerebellar, intracerebroventricular, intracolic, intracervical, intragastric, intrahepatic, intramyocardial, intraosteal, intrapelvic, intrapericardiac, intraperitoneal, intrapleural, intraprostatic, intrapulmonary, intrarectal, intrarenal, intraretinal, intraspinal, intrasynovial, intrathoracic, intrauterine, intravesical, intralesional, bolus, vaginal, rectal, buccal, sublingual, intranasal, or transdermal means. An dAb, specified portion or variant compositions can be prepared for use for parenteral (subcutaneous, intramuscular, intravenous, intrarticular, etc.) administration particularly in the form of liquid solutions or suspensions; for use in topical, vaginal or rectal administration particularly in semisolid forms, such as creams and suppositories; for buccal, or sublingual administration particularly in the form of tablets or capsules; or intranasally particularly in the form of powders, nasal drops or aerosols or certain agents; or transdermally particularly in the form of a gel, ointment, lotion, suspension or patch delivery system with chemical enhancers, such as dimethyl sulfoxide, to either modify the skin structure or to increase the drug concentration in the transdermal patch (Junginger et al., In “Drug Permeation Enhancement”; Hsieh, D. S., Eds., pp. 59-90 (Marcel Dekker, Inc. New York 1994, entirely incorporated herein by reference), or with oxidising agents that enable the application of formulations containing proteins and peptides onto the skin (WO 98/53847), or applications of electric fields to create transient transport pathways, such as electroporation, or to increase the mobility of charged drugs through the skin such as iontophoresis, or application of ultrasound, such as sonophoresis (U.S. Pat. Nos. 4,309,989 and 4,767,402; the above publications and patents being entirely incorporated herein by reference).


For pulmonary administration, preferably, at least one dAb, specified portion or variant composition is delivered in a particle size effective for reaching the lower airways of the lung or sinuses. According to the invention, at least one dAb, specified portion or variant can be delivered by any of a variety of inhalation or nasal devices known in the art for administration of a therapeutic agent by inhalation. These devices capable of depositing aerosolised formulations in the sinus cavity or alveoli of a patient include metered dose inhalers, nebulisers, dry powder generators, sprayers, and the like. Other devices suitable for directing the pulmonary or nasal administration of a dAb, specified portion or variants thereof are also known in the art. All such devices can use formulations suitable for the administration for the dispensing of a dAb, specified portion or variant in an aerosol. Such aerosols can be comprised of either solutions (both aqueous and non aqueous) or solid particles. Metered dose inhalers like the Ventolin® metered dose inhaler, typically use a propellent gas and require actuation during inspiration (See, e.g., WO 94/16970, WO 98/35888). Dry powder inhalers like Turbuhaler™ (Astra), Rotahaler® (Glaxo), Diskus® (Glaxo), Spiros™ inhaler (Dura), devices marketed by Inhale Therapeutics, and the Spinhaler® powder inhaler (Fisons), use breath-actuation of a mixed powder (U.S. Pat. No. 4,668,218 Astra, EP 237507 Astra, WO 97/25086 Glaxo, WO 94/08552 Dura, U.S. Pat. No. 5,458,135 Inhale, WO 94/06498 Fisons, entirely incorporated herein by reference). Nebulisers like AERx™ Aradigm, the Ultravent® nebuliser (Mallinckrodt), and the Acorn II® nebuliser (Marquest Medical Products; U.S. Pat. No. 5,404,871 Aradigm, WO 97/22376), the above references entirely incorporated herein by reference, produce aerosols from solutions, while metered dose inhalers, dry powder inhalers, etc. generate small particle aerosols. These specific examples of commercially available inhalation devices are intended to be representative of specific devices suitable for the practice of this invention, and are not intended as limiting the scope of the invention. Preferably, a composition comprising at least one dAb or specified portion or variant is delivered by a dry powder inhaler or a sprayer. There are several desirable features of an inhalation device for administering at least one dAb, specified portion or variant of the present invention. For example, delivery by the inhalation device is advantageously reliable, reproducible, and accurate. The inhalation device can optionally deliver small dry particles, e.g. less than about 10 μm, preferably about 1-5 μm, for good respirability.


Formulations for oral administration of at least one dAb, specified portion or variant, rely on the co-administration of adjuvants (e.g., resorcinols and nonionic surfactants, such as polyoxyethylene oleyl ether and n-hexadecylpolyethylene ether) to increase artificially the permeability of the intestinal walls, as well as the co-administration of enzymatic inhibitors (e.g., pancreatic trypsin inhibitors, diisopropylfluorophosphate (DFF) and trasylol) to inhibit enzymatic degradation. The active constituent compound of the solid-type dosage form for oral administration can be mixed with at least one additive, including sucrose, lactose, cellulose, mannitol, trehalose, raffinose, maltitol, dextran, starches, agar, arginates, chitins, chitosans, pectins, gum tragacanth, gum arabic, gelatin, collagen, casein, albumin, synthetic or semisynthetic polymer, and glyceride. These dosage forms can also contain other type(s) of additives, e.g., inactive diluting agent, lubricant, such as magnesium stearate, paraben, preserving agent, such as sorbic acid, ascorbic acid, α-tocopherol, antioxidant, such as cysteine, disintegrator, binder, thickener, buffering agent, sweetening agent, flavoring agent, perfuming agent, etc.


Tablets and pills can be further processed into enteric-coated preparations. The liquid preparations for oral administration include emulsion, syrup, elixir, suspension and solution preparations allowable for medical use. These preparations may contain inactive diluting agents ordinarily used in said field, e.g., water. Liposomes have also been described as drug delivery systems for insulin and heparin (U.S. Pat. No. 4,239,754). More recently, microspheres of artificial polymers of mixed amino acids (proteinoids) have been used to deliver pharmaceuticals (U.S. Pat. No. 4,925,673). Furthermore, carrier compounds described in U.S. Pat. Nos. 5,879,681 and 5,871,753 are used to deliver biologically active agents orally and are known in the art.


For absorption through mucosal surfaces, compositions and methods of administering at least one dAb, specified portion or variant include an emulsion comprising a plurality of submicron particles, a mucoadhesive macromolecule, a bioactive peptide, and an aqueous continuous phase, which promotes absorption through mucosal surfaces by achieving mucoadhesion of the emulsion particles (U.S. Pat. No. 5,514,670). Mucus surfaces suitable for application of the emulsions of the present invention can include corneal, conjunctival, buccal, sublingual, nasal, vaginal, pulmonary, stomachic, intestinal, and rectal routes of administration. Formulations for vaginal or rectal administration, e.g. suppositories, can contain as excipients, for example, polyalkyleneglycols, vaseline, cocoa butter, and the like. Formulations for intranasal administration can be solid and contain as excipients, for example, lactose or can be aqueous or oily solutions of nasal drops. For buccal administration, excipients include sugars, calcium stearate, magnesium stearate, pregelinatined starch, and the like (U.S. Pat. No. 5,849,695).


For transdermal administration, the at least one dAb, specified portion or variant is encapsulated in a delivery device, such as liposomes or polymeric nanoparticles, microparticles, microcapsules, or microspheres (referred to collectively as microparticles unless otherwise stated). A number of suitable devices are known, including microparticles made of synthetic polymers, such as polyhydroxy acids, such as polylactic acid, polyglycolic acid and copolymers thereof, polyorthoesters, polyanhydrides, and polyphosphazenes, and natural polymers, such as collagen, polyamino acids, albumin and other proteins, alginate and other polysaccharides, and combinations thereof (U.S. Pat. No. 5,814,599).


It can be sometimes desirable to deliver the domain antibodies of the present invention to the subject over prolonged periods of time, for example, for periods of one week to one year from a single administration. Various slow release, depot or implant dosage forms can be utilised. For example, a dosage form can contain a pharmaceutically acceptable non-toxic salt of the compounds that has a low degree of solubility in body fluids, for example, (a) an acid addition salt with a polybasic acid, such as phosphoric acid, sulfuric acid, citric acid, tartaric acid, tannic acid, pamoic acid, alginic acid, polyglutamic acid, naphthalene mono- or di-sulfonic acids, polygalacturonic acid, and the like; (b) a salt with a polyvalent metal cation, such as zinc, calcium, bismuth, barium, magnesium, aluminium, copper, cobalt, nickel, cadmium and the like, or with an organic cation formed from e.g., N,N′-dibenzyl-ethylenediamine or ethylenediamine; or (c) combinations of (a) and (b) e.g., a zinc tannate salt. Additionally, the compounds of the present invention or, preferably, a relatively insoluble salt, such as those just described, can be formulated in a gel, for example, an aluminium monostearate gel with, e.g. sesame oil, suitable for injection. Particularly preferred salts are zinc salts, zinc tannate salts, pamoate salts, and the like. Another type of slow release depot formulation for injection would contain the compound or salt dispersed for encapsulation in a slow degrading, non-toxic, non-antigenic polymer, such as a polylactic acid/polyglycolic acid polymer, for example, as described in U.S. Pat. No. 3,773,919. The compounds or, preferably, relatively insoluble salts, such as those described above, can also be formulated in cholesterol matrix silastic pellets, particularly for use in animals. Additional slow release, depot or implant formulations, e.g., gas or liquid liposomes, are known in the literature (U.S. Pat. No. 5,770,222 and Robinson Ed., 1978).


All scientific citations, patents, patent applications and manufacturer's technical specifications referred to hereinafter are incorporated herein by reference in their entirety.


Throughout this specification, unless the context requires otherwise, the word “comprise”, or variations such as “comprises” or “comprising”, will be understood to imply the inclusion of a stated element or integer or group of elements or integers but not the exclusion of any other element or integer or group of elements or integers.


It is to be understood that unless otherwise indicated, the subject invention is not limited to specific formulation components, manufacturing methods, dosage regimens, or the like, as such may vary. It is also to be understood that the terminology used herein is for the purpose of describing particular embodiments only and is not intended to be limiting.


It must be noted that, as used in the subject specification, the singular forms “a”, “an” and “the” include plural aspects unless the context clearly dictates otherwise. Thus, for example, reference to “a target” includes a single target, as well as two or more targets; reference to “an oligodendrocyte” includes a single oligodendrocyte, as well as two or more oligodendrocytes; reference to “the formulation” includes a single formulation, as well as two or more formulations; and so forth.


The present invention is further described by the following non-limiting Examples.


Example 1
Identification of Key Framework Variants
Design of the Framework Variants

A directed mutagenesis approach was taken in order to identify key framework residues in a domain antibody construct that improved its function. Protein functions that were improved were neutralization potency, antigen binding efficiency and/or expression levels.


The unmodified domain antibody construct comprises a variable light (VL) domain antibody (dAb) targeted to human TNFα, a modified hinge region and the heavy chain constant region of human IgG1 but wherein the constant region has a truncated CH1 domain (WO 2007/087673). The unmodified dAb construct will herein be referred to as ‘TNFα dAb Fc’. TNFα dAb Fc has the sequence of SEQ. ID. NO. 2


Complementarity-determining regions (CDRs) and framework residues of the VL domain of the domain antibody Fc construct, or TNFα dAb Fc, were identified using definitions and consensus sequences of the Kabat convention (Kabat et al., “Sequence of Proteins of Immunological Interest”, US Department of Health and Human Services).


Single substitution mutations were made to the framework regions of the VL domain of TNFα dAb Fc construct. Each ‘wild-type’ residue was substituted to one of nine amino acids. The selected amino acids are representative of the major side-chain chemistries provided by the twenty amino acids. The nine amino acids and their type of side-chain functionality are:


alanine (A), small;


serine (S) and histidine (H), nucleophilic;


glutamine (Q), amide;


aspartic acid (D), acidic;


lysine (K), basic;


leucine (L) and proline (P), hydrophobic; and


tyrosine (Y), aromatic.


Residues within and some immediately adjacent to the CDRs of the VL domain were not mutated. In cases where the wild-type residue is one of the nine representative amino acids, then substitutions into the remaining eight residues were made. In total, 78 framework residues were mutated and 640 variants containing single substitution mutations were generated in VL domains of the TNFα dAb Fc construct.


The gene fragments encoding the variants were optimized for expression in mammalian cells. They were assembled from synthetic oligonucleotides and/or PCR products and cloned into a vector suitable for mammalian cell expression. The final DNA constructs were verified by sequencing.


First Round Screen Using TNFα Binding ELISA

The framework variants containing single substitution mutations, as well as the unmodified TNFα dAb Fc construct, were assessed for their ability to bind to target antigen, human TNFα, in a TNFα binding ELISA.


The variants were transiently expressed using suspension variant of the CHOK1 cell line. In this example, small-scale transient transfections were performed using the Freestyle™ MAX CHO transfection method (Invitrogen; 2 ml transfections in 24-well plate format). Six days after transfection, conditioned medium of transfected cells was harvested by centrifugation and tested in the following ELISA.


TNFα Binding ELISA

Ninety-six well flat-bottom plates (MaxiSorp™, Nunc™) were coated with recombinant human TNFα (CytoLab; 1 μg/ml) in carbonate coating buffer (35 mM sodium carbonate, 15 mM sodium hydrogen carbonate, pH 9.6) overnight at 4° C. The plates were washed three times in PBS containing 0.05% Tween-20 (PBS-T) then PBS. The plates were incubated in blocking solution (1% w/v BSA diluted in PBS) for 2 hours at room temperature. After washing three times in PBS-T and PBS, 50 μl of the conditioned medium and 50 μl of diluent buffer (1% w/v BSA diluted in PBS) was added to the wells and incubated for 2 hours at room temperature. The plates were washed three times in PBS-T and PBS then incubated with secondary antibody HRP goat anti-human IgG (H+L; Zymed®, diluted 1:2000 with diluent buffer). After one hour incubation at room temperature, plates were washed three times as above then 100 μl of 3,3′,5,5′tetramethylbenzidine (TMB; Sigma Aldrich®) solution added to the wells. Colorimetric development was performed at room temperature and stopped with 50 μl of 1 M hydrochloric acid and absorbance read at 450 nm.



FIG. 1 is a representative graph of TNFα binding ELISA showing resultant binding of framework variants with substitutions at position forty-nine. Table 1 lists the ELISA results (A 450 nm values) for the 640 variants screened.









TABLE 1







TNFα binding ELISA data of the 640 single substitution


TNFα dAb framework variants.










TNFα dAb




variants
A450 (AU)














D1A
0.978



D1S
0.738



D1L
0.762



D1Y
0.971



D1Q
0.683



D1H
0.740



D1P
0.496



D1K
0.875



I2A
0.917



I2S
0.549



I2L
0.751



I2Y
0.876



I2D
0.616



I2Q
0.459



I2H
0.531



I2P
0.655



I2K
0.550



Q3A
1.057



Q3S
0.664



Q3L
0.610



Q3Y
0.963



Q3D
0.709



Q3H
0.692



Q3P
0.375



Q3K
0.589



M4A
0.289



M4S
0.230



M4L
0.731



M4Y
0.358



M4D
0.246



M4Q
0.335



M4H
0.335



M4P
0.296



M4K
0.410



T5A
0.973



T5L
0.661



T5Y
0.969



T5D
0.648



T5Q
0.685



T5H
0.703



T5P
0.333



T5K
0.643



Q6A
0.973



Q6S
0.544



Q6L
0.324



Q6Y
0.969



Q6D
0.252



Q6H
0.380



Q6P
0.262



Q6K
0.304



S7A
1.016



S7L
0.749



S7Y
0.975



S7D
0.872



S7Q
0.781



S7H
0.743



S7P
0.310



S7K
0.769



P8A
0.892



P8S
0.680



P8L
0.658



P8Y
0.694



P8D
0.564



P8Q
0.731



P8H
0.627



P8K
0.623



S9A
0.981



S9L
0.972



S9Y
0.769



S9D
0.973



S9Q
0.768



S9H
0.850



S9P
0.838



S9K
0.783



S10A
0.997



S10L
0.988



S10Y
0.795



S10D
0.813



S10Q
0.980



S10H
0.764



S10P
0.687



S10K
0.803



L11A
0.912



L11S
0.858



L11Y
0.708



L11D
0.299



L11Q
1.102



L11H
0.650



L11P
0.279



L11K
0.556



S12A
0.939



S12L
0.710



S12Y
0.704



S12D
0.636



S12Q
0.762



S12H
0.699



S12P
0.911



S12K
0.714



A13S
0.735



A13L
0.879



A13Y
0.628



A13D
0.229



A13Q
0.648



A13H
0.699



A13P
0.685



A13K
0.723



S14A
0.966



S14L
0.659



S14Y
0.937



S14D
0.787



S14Q
0.752



S14H
0.742



S14P
0.649



S14K
0.782



V15A
0.912



V15S
0.724



V15Y
0.737



V15D
0.561



V15Q
0.557



V15H
0.780



V15P
0.965



V15K
0.740



G16S
0.619



G16L
0.633



G16Y
0.942



G16D
0.542



G16Q
0.594



G16H
0.677



G16P
0.156



G16K
0.896



D17A
0.875



D17S
1.052



D17L
1.000



D17Y
1.083



D17Q
0.903



D17H
1.077



D17P
0.953



D17K
0.295



R18A
0.894



R18S
1.141



R18L
0.957



R18Y
0.900



R18D
0.984



R18Q
0.982



R18H
1.027



R18P
0.855



R18K
0.867



V19A
0.923



V19S
0.643



V19Y
0.254



V19D
0.157



V19Q
0.223



V19H
0.560



V19P
0.341



V19K
0.750



T20A
0.864



T20L
1.034



T20Y
1.119



T20D
1.043



T20Q
1.023



T20H
0.981



T20P
0.881



T20K
1.017



I21A
0.405



I21S
0.263



I21L
0.935



I21Y
0.191



I21D
0.165



I21Q
0.143



I21H
0.173



I21P
0.161



I21K
0.194



T22A
0.941



T22L
1.010



T22Y
1.036



T22D
0.985



T22Q
1.070



T22H
1.088



T22P
0.645



T22K
1.062



Y36A
0.185



Y36S
0.263



Y36L
0.237



Y36D
0.168



Y36Q
0.167



Y36H
0.149



Y36P
0.243



Y36K
0.378



Q37A
0.877



Q37S
0.962



Q37L
0.885



Q37Y
0.549



Q37D
0.251



Q37H
0.947



Q37P
0.294



Q37K
1.042



Q38A
0.783



Q38S
0.992



Q38L
0.649



Q38Y
0.793



Q38D
0.178



Q38H
1.101



Q38P
0.156



Q38K
0.909



K39A
0.802



K39S
1.004



K39L
1.056



K39Y
1.019



K39D
0.754



K39Q
0.982



K39H
1.062



K39P
0.949



P40A
0.901



P40S
1.031



P40L
1.003



P40Y
1.048



P40D
1.090



P40Q
1.053



P40H
0.990



P40K
1.041



G41S
1.062



G41L
0.906



G41Y
0.929



G41D
0.883



G41Q
1.066



G41H
0.925



G41P
0.966



G41K
0.877



K42A
0.872



K42S
1.092



K42L
0.816



K42Y
1.013



K42D
1.034



K42Q
0.903



K42H
1.074



K42P
0.246



A43S
0.831



A43L
0.718



A43Y
0.976



A43D
0.661



A43Q
1.051



A43H
1.018



A43P
0.825



A43K
1.024



P44A
0.261



P44S
0.247



P44L
1.111



P44Y
0.339



P44D
0.163



P44Q
0.182



P44H
0.309



P44K
0.187



K45A
0.865



K45S
1.038



K45L
1.112



K45Y
1.104



K45D
1.194



K45Q
1.063



K45H
1.175



K45P
0.191



L46A
0.185



L46S
0.171



L46Y
0.256



L46D
0.191



L46Q
0.234



L46H
0.166



L46P
0.179



L46K
0.243



L47A
0.257



L47S
0.222



L47Y
0.903



L47D
0.215



L47Q
0.369



L47H
0.284



L47P
0.193



L47K
0.271



I48A
1.248



I48S
0.293



I48Y
0.408



I48D
0.239



I48Q
0.265



I48H
0.234



I48P
0.193



I48K
0.253



Y49A
0.455



Y49S
1.154



Y49L
0.592



Y49D
1.233



Y49Q
0.242



Y49H
1.121



Y49P
0.188



Y49K
0.174



G57A
1.181



G57S
1.077



G57L
1.073



G57Y
1.084



G57D
0.473



G57Q
1.113



G57H
1.186



G57P
0.496



G57K
1.191



V58A
1.021



V58S
0.399



V58Y
0.247



V58D
0.185



V58Q
0.207



V58H
0.220



V58P
0.255



V58K
0.205



P59A
0.219



P59S
1.121



P59L
0.901



P59Y
0.893



P59D
1.123



P59Q
1.052



P59H
1.263



P59K
0.691



S60A
1.366



S60L
1.212



S60Y
1.352



S60D
1.208



S60Q
1.125



S60H
1.486



S60P
1.337



S60K
1.459



R61A
0.210



R61S
0.768



R61L
0.211



R61Y
0.783



R61D
0.215



R61Q
0.395



R61H
1.365



R61P
0.225



F62A
0.220



F62S
0.206



F62L
0.613



F62Y
0.960



F62D
0.208



F62Q
0.184



F62H
0.241



F62P
0.226



F62K
0.292



S63A
0.897



S63L
0.751



S63Y
0.923



S63D
0.873



S63Q
0.964



S63H
0.774



S63P
0.205



S63K
0.997



G64S
0.907



G64L
0.189



G64Y
0.193



G64D
0.171



G64Q
0.163



G64H
0.263



G64P
0.643



G64K
0.226



S65A
0.903



S65L
0.924



S65Y
1.010



S65D
0.593



S65Q
0.889



S65H
1.052



S65P
0.190



S65K
1.062



G66S
0.393



G66L
0.842



G66Y
0.909



G66D
0.808



G66Q
0.972



G66H
0.883



G66P
0.184



G66K
0.968



S67A
0.944



S67L
0.863



S67Y
0.944



S67D
1.024



S67Q
0.419



S67H
0.999



S67P
0.188



S67K
0.819



G68S
1.010



G68L
0.443



G68Y
0.305



G68D
1.002



G68Q
0.889



G68H
0.325



G68P
0.827



G68K
0.587



T69A
0.916



T69L
1.040



T69Y
1.034



T69D
1.205



T69Q
1.018



T69H
0.904



T69P
0.760



T69K
1.037



D70A
0.916



D70S
1.016



D70L
0.991



D70Y
0.938



D70Q
1.118



D70H
1.027



D70P
0.349



D70K
0.977



F71A
0.185



F71S
0.226



F71L
0.208



F71Y
0.597



F71D
0.176



F71Q
0.191



F71H
0.315



F71P
0.205



F71K
0.213



T72A
0.905



T72L
1.037



T72Y
0.193



T72D
0.916



T72Q
0.966



T72H
1.026



T72P
0.149



T72K
0.936



L73A
0.225



L73S
0.152



L73Y
0.204



L73D
0.172



L73Q
0.155



L73H
0.155



L73P
0.173



L73K
0.269



T74A
0.906



T74L
0.718



T74Y
0.640



T74D
0.962



T74Q
0.884



T74H
0.929



T74P
0.187



T74K
0.905



I75A
0.196



I75S
0.194



I75L
0.858



I75Y
0.168



I75D
0.167



I75Q
0.203



I75H
0.201



I75P
0.183



I75K
0.180



S76A
0.970



S76L
0.832



S76Y
0.797



S76D
0.758



S76Q
0.761



S76H
0.902



S76P
0.472



S76K
0.767



S77A
0.925



S77L
0.761



S77Y
0.852



S77D
0.485



S77Q
0.723



S77H
0.827



S77P
0.952



S77K
0.800



L78A
0.847



L78S
0.610



L78Y
0.379



L78D
0.176



L78Q
0.273



L78H
0.210



L78P
0.369



L78K
0.489



L79V
1.435



L79P
0.246



L79A
1.382



L79G
1.404



L79S
1.392



L79Y
1.408



L79D
1.612



L79K
1.259



L79Q
1.325



L79H
1.300



P80A
0.947



P80S
0.910



P80L
0.794



P80Y
0.766



P80D
0.741



P80Q
0.764



P80H
0.770



P80K
0.782



E81A
0.896



E81S
0.787



E81L
0.824



E81Y
0.793



E81D
0.799



E81Q
0.808



E81H
0.795



E81P
0.761



E81K
0.803



D82A
0.219



D82S
0.224



D82L
0.244



D82Y
0.265



D82Q
0.195



D82H
0.251



D82P
0.204



D82K
0.346



F83A
1.144



F83G
1.052



F83S
0.503



F83L
1.002



F83Y
1.291



F83D
1.141



F83Q
1.022



F83H
1.022



F83P
0.453



A84S
0.645



A84L
0.212



A84Y
0.179



A84D
0.177



A84Q
0.280



A84H
0.326



A84P
0.178



A84K
0.292



T85A
0.920



T85L
0.742



T85Y
0.895



T85D
0.590



T85Q
0.764



T85H
0.769



T85P
0.195



T85K
0.682



Y86A
0.153



Y86S
0.180



Y86L
0.205



Y86D
0.194



Y86Q
0.204



Y86H
0.212



Y86P
0.182



Y86K
0.185



Y87A
0.219



Y87S
0.201



Y87L
0.717



Y87D
0.208



Y87Q
0.192



Y87H
0.222



Y87P
0.176



Y87K
0.283



F98A
0.199



F98S
0.160



F98L
0.196



F98Y
0.666



F98D
0.179



F98Q
0.169



F98H
0.189



F98P
0.178



F98K
0.180



G99S
0.577



G99L
0.192



G99Y
0.213



G99D
0.177



G99Q
0.181



G99H
0.190



G99P
0.181



G99K
0.207



Q100A
0.876



Q100S
0.815



Q100L
0.628



Q100Y
0.644



Q100D
0.318



Q100H
0.729



Q100P
0.775



Q100K
0.376



G101S
0.195



G101L
0.191



G101Y
0.192



G101D
0.156



G101Q
0.199



G101H
0.214



G101P
0.961



G101K
0.295



T102A
0.383



T102L
0.210



T102Y
0.183



T102D
0.154



T102Q
0.204



T102H
0.274



T102P
0.169



T102K
0.166



K103A
0.916



K103S
1.024



K103L
0.816



K103Y
0.910



K103D
0.972



K103Q
0.258



K103H
0.975



K103P
0.319



V104A
0.667



V104S
0.208



V104Y
0.229



V104D
0.147



V104Q
0.183



V104H
0.157



V104P
0.182



V104K
0.274



E105A
0.869



E105S
0.254



E105L
0.688



E105Y
0.382



E105D
0.204



E105Q
0.412



E105H
0.506



E105P
0.237



E105K
0.612



I106A
0.867



I106S
0.172



I106L
0.165



I106Y
0.336



I106D
0.402



I106Q
0.164



I106H
0.306



I106P
0.203



I106K
0.501



K106AA
0.876



K106AS
0.294



K106AL
0.152



K106AY
0.270



K106AD
0.374



K106AQ
0.139



K106AH
0.151



K106AP
0.598



R107A
0.972



R107S
0.530



R107L
0.389



R107Y
0.395



R107D
0.161



R107Q
0.365



R107H
0.468



R107P
0.531



R107K
0.449



TNFα dAb Fc
0.819










This first round screen using TNFα binding ELISA identified framework variants where single substitution mutations were tolerated, i.e. variants that were expressed, secreted and retained their ability to bind to target antigen, human TNFα.


Second Round Testing of Variants for Neutralization of TNFα Induced Cytotoxicity

A subset of TNFα dAb variants were selected and assessed for their potency in neutralizing TNFα-mediated cytotoxicity in the murine L929 cell line.


Medium-scale transfections (30 ml) were performed to obtain sufficient protein amounts for the L929 assay. The variants were transiently expressed in suspension variant of CHOK1 cell line using the Freestyle™ MAX CHO transfection method. Six days after transfection, conditioned media was harvested from the transfected cells by centrifugation and the supernatants filtered using 0.22 μm membrane filter. The TNFα dAb variants were purified by Protein A affinity chromatography. After sample loading, unbound proteins were washed off using PBS, variants were eluted using 0.1 M citric acid pH 3.5 and fractions neutralized with the addition of 1 M Tris (pH 9.0). The variants were concentrated and buffer exchanged into PBS using sample concentrators (e.g. Microcon®YM-30 units; Millipore®). Protein concentration was determined by bicinchoninic acid assay (BCA®; Pierce®).


High Throughput 5-Point L929 Neutralization Assay

Serial half log dilutions of TNFα dAb variants in RPMI media (containing 10% foetal bovine serum and 2 mM L-glutamine) were prepared in 50 μl volume ranging from 6.25 μg/ml to 0.124 μg/ml across five wells in 96-well flat bottom plates. To each of the test wells, 25 μl of human TNFα (1.5 ng/ml), 25 μl actinomycin D (40 μg/ml) and 50 μl L929 cells (5×105 cells/ml) were added. Controls included a TNFα standard curve ranging from 3125 pg/ml to 0.172 pg/ml across eleven wells, wells containing no TNFα (100% viability) and no cells (background). Assay plates were incubated at 37° C. in a 5% CO2 humidified incubator for 20 hours then a further 2 hours after the addition of 30 μl 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-terazolium (MTS)/phenazine ethosulfate (PES). Absorbance was read at 492 nm and viability curves calculated using the average absorbance of duplicate wells. Viability curves were fitted to a sigmoidal dose model using GraphPad software (Prism®).



FIG. 2 is a representative result generated by the high throughput 5-point L929 assay and Table 2 presents the EC50 values of the tested TNFα dAb Fc framework variants.









TABLE 2







TNFα neutralization potency data of single substitution TNFα


dAb framework variants. Neutralization of TNFα-induced cytotoxicity


results measured as EC50 values in the L929 assay is listed. Lower


values indicate improved potency. This is an exception for those


variants where the resulting curve is a straight line as shown in


FIG. 2 for variant L11P. This data was obtained using the high throughput


5-point format of the L929 neutralization assay.










TNFα dAb
Standardized



variants
EC50 (μg/ml)














D1A
0.556



D1S
0.846



D1L
6.038



D1Y
0.676



D1Q
1.097



D1H
1.128



D1K
0.833



I2A
1.232



I2S
2.789



I2L
0.593



I2Y
3.606



I2D
3479.239



I2H
80.713



I2P
0.725



I2K
2.789



Q3A
2.323



Q3S
2.580



Q3L
6.443



Q3Y
0.450



Q3D
0.809



Q3H
0.795



Q3P
20.462



Q3K
1.819



Q6A
123.057



S7A
0.383



S7L
1.636



S7Y
0.928



S7D
0.676



S7Q
0.488



S7H
1.094



S7K
2.104



S9A
0.671



S9L
0.980



S9Y
1.996



S9D
0.880



S9Q
1.167



S9H
1.226



S9P
1.299



S9K
1.229



S10A
0.661



S10L
0.616



S10Y
1.760



S10Q
1.419



S10H
1.284



S10P
6.586



S10K
2.718



L11A
0.912



L11S
1.643



L11Y
2.774



L11D
10.150



L11Q
1.013



L11P
0.000



L11K
9.470



S12A
0.796



S12L
0.252



S12Y
1.066



S12D
1.765



S12Q
0.822



S12H
1.443



S12P
1.352



S12K
1.046



S14A
0.681



S14L
2.665



S14Y
1.924



S14D
2.262



S14Q
0.841



S14H
4134.019



S14P
9.960



S14K
1.760



V15A
2.403



V15P
1.243



G16Y
1.972



D17A
4.668



D17S
0.909



D17L
2.659



D17Q
0.863



D17H
10.542



D17P
667.543



D17K
8.844



R18A
3.208



R18S
0.851



R18L
1.000



R18Y
1.588



R18D
0.741



R18Q
2.749



R18H
2.387



R18P
1.191



R18K
0.957



V19A
1.856



T20A
1.597



T20L
1.933



T20Y
6.962



T20D
3.221



T20Q
2.020



T20H
1.686



T20K
4.381



I21L
2.395



T22A
0.725



T22L
2.509



T22Y
2.853



T22D
1.430



T22Q
2.258



T22H
11.548



T22K
2.001



Q37S
1.978



Q37H
16.398



Q37K
6.567



Q38A
9.108



Q38S
1.837



Q38Y
5045.567



Q38H
1.010



Q38K
1.890



K39S
1.601



K39Q
0.000



P40A
1.646



P40S
2.855



P40L
5.701



P40Y
26.688



P40D
1.644



P40Q
5.350



P40H
3.291



P40K
5.286



G41S
5.343



G41Y
3.569



G41Q
2.386



G41H
2.414



G41P
7.333



K42A
0.723



K42S
0.995



K42L
1.225



K42Y
3.346



K42D
1.496



K42Q
1.059



K42H
2.260



A43Y
666.360



A43Q
2.686



A43H
2.742



A43K
3.130



P44A
24.381



P44S
28.760



P44L
6.193



P44Y
27.549



P44D
802.274



P44Q
0.000



P44K
0.000



K45A
2.212



K45S
0.537



K45L
1.272



K45Y
1.377



K45D
1.404



K45Q
4.415



K45H
1.767



Y49A
6.563



Y49S
13.159



Y49L
15.836



Y49D
1.540



Y49Q
590.486



Y49H
0.731



Y49P
184.975



Y49K
1573.382



G57A
2.061



G57S
0.618



G57L
2.821



G57Y
1.005



G57D
0.948



G57Q
0.761



G57H
0.795



G57P
6.526



G57K
1.030



P59A
0.452



P59S
0.689



P59L
2.247



P59Y
1.764



P59D
0.697



P59Q
1.172



P59H
0.971



P59K
7.043



S60A
0.176



S60L
0.640



S60D
0.417



S60Q
0.746



S60H
0.735



S60P
0.895



S60K
0.867



R61D
0.000



R61H
8.868



F62A
0.000



F62S
0.000



F62Y
0.694



F62D
0.000



F62H
12.329



F62P
0.000



S63Y
2.025



S63Q
1.331



S63K
1.335



G64S
2.348



S65L
1.010



S65Y
4.725



S65H
3.713



S65K
3.493



G66Y
2.438



G66Q
2166.311



G66K
541.923



S67D
5.133



G68S
2.543



G68D
6.361



T69A
19.621



T69L
3.103



T69Y
2.490



T69D
1.345



T69Q
2.852



T69K
8.984



D70A
2.039



D70S
1.584



D70Q
3.830



T72H
5.469



L79A
1.351



L79S
0.378



L79Y
0.392



L79D
0.766



L79H
1.343



L79K
0.503



F83A
0.682



F83S
0.839



F83L
0.636



F83Y
0.493



F83D
0.947



F83Q
1.036



F83H
1.332



F83P
16.898



F83G
6.209



K103S
1.887



TNFα dAb Fc
1.200










The mutagenesis approach taken in the present invention identified hotspots, as well as specific substitutions, in the framework regions that resulted in increased potency of the TNFα dAb Fc variants for the TNFα antigen.


Example 2
TNFα dAb Variants Containing Multiple Substitutions

TNFα dAb Fc variants containing multiple substitutions in the VL domain were tested to determine whether the improved potency imparted by the single substitution variants can be combined in an additive manner. To test this, a subset of six single substitutions was selected and a panel of TNFα dAb Fc variants containing double, triple, quadruple substitutions, including a variant containing six substitution mutations were produced. The six selected substitutions are Q3Y, S7Q, S12L, S60A, L79Y and F83Y. The variants comprising multiple substitutions were produced in the same manner as the unmodified TNFα dAb Fc protein, as described in Example 1. That is, a domain antibody construct comprising a variable light (VL) domain antibody (dAb) targeted to human TNFα, a modified hinge region and the heavy chain constant region of human IgG1 but wherein the constant region has a truncated CH1 domain (WO 2007/087673). The sequence of this modified Fc is set out in SEQ ID NO. 6. The constructs described in this Example are herein collectively referred to as ‘Combination TNFα dAb framework variants’.


The panel of combination TNFα dAb framework variants were produced as described above. Briefly, gene fragments encoding the selected variants were synthesized and inserted into a vector suitable for mammalian cell expression. The variants were transiently expressed in suspension variant of the CHOK1 cell line using the Freestyle™ MAX CHO transfection method. The conditioned media was subjected to Protein A affinity chromatography, and the purified combination variants quantitated using the BCA® assay.


The combination variants were assayed for their ability to neutralize TNFα-mediated cytotoxicity using the standard 11-point format of the L929 neutralization assay.


Standard 11-Point L929 Neutralization Assay

This assay was performed in a similar manner as described in ‘high throughput 5-point L929 neutralization assay’ (see Example 1). In this format serial half log dilutions of combination variants in RPMI media were prepared in 50 μl volume ranging from 52.5 μg/ml to 0.0005 μg/ml across eleven wells in 96-well flat bottom plates.


Table 3 presents the EC50 values of the combination TNFα dAb framework variants. Several combination variants exhibited further increases in potency, i.e. the ability to neutralize TNFα, when compared to the individual single substitution framework variants.









TABLE 3







TNFα neutralization potency data of combination TNFα


dAb framework variants, as measured by neutralization of TNFα-induced


cytotoxicity of L929 cells. Also listed are additional single substituted


TNFα dAb variants incorporating other amino acids of the same


side-chain functionalities. These are data obtained using the standard


11-point format of the L929 neutralization assay.









Standardized EC50 (μg/ml)


Variants
Average ± S.D.





Q3Y
1.198 ± 0.345


Q3F
0.910 ± 0.007


S7Q
1.496 ± 0.320


S7G
0.871 ± 0.220


S7N
0.673 ± 0.139


S12L
0.922 ± 0.269


S12V
0.979 ± 0.079


S12T
0.769 ± 0.187


S60A
0.896 ± 0.241


S60E
1.144 ± 0.183


S60G
0.644 ± 0.171


L79Y
0.950 ± 0.304


L79F
1.027 ± 0.063


L79G
0.549 ± 0.041


F83Y
0.990 ± 0.300


Q3Y_S7Q
0.922 ± 0.271


Q3Y_S12L
0.705 ± 0.011


Q3Y_S60A
0.968 ± 0.080


Q3Y_L79Y
1.205 ± 0.108


Q3Y_F83Y
2.961 ± 0.062


S7Q_S12L
0.877 ± 0.661


S7Q_S60A
0.852 ± 0.266


S7Q_L79Y
1.594 ± 0.856


S7Q_F83Y
2.240 ± 0.023


S12L_S60A
2.079 ± 0.448


S12L_F83Y
1.735 ± 0.374


S60A_F83Y
0.583 ± 0.196


L79Y_F83Y
1.292 ± 0.555


Q3Y_S7Q_S12L
1.092 ± 0.298


Q3Y_S7Q_S60A
1.431 ± 0.096


Q3Y_S7Q_L79Y
4.376 ± 3.002


Q3Y_S7Q_F83Y
2.313 ± 0.607


Q3Y_S12L_S60A
3.334 ± 1.040


Q3Y_S12L_L79Y
1.141 ± 0.125


Q3Y_S12L_F83Y
3.479 ± 0.681


Q3Y_S60A_F83Y
1.039 ± 0.441


Q3Y_L79Y_F83Y
2.408 ± 0.510


S7Q_S12L_S60A
3.539 ± 0.448


S7Q_S12L_L79Y
2.280 ± 0.078


S7Q_S12L_F83Y
2.887 ± 2.004


S7Q_S60A_L79Y
2.131 ± 0.241


S7Q_S60A_F83Y
2.243 ± 0.455


S7Q_L79Y_F83Y
1.016 ± 0.245


S12L_S60A_L79Y
1.481 ± 0.364


S12L_S60A_F83Y
0.971 ± 0.506


S12L_L79Y_F83Y
0.959 ± 0.057


S60A_L79Y_F83Y
1.367 ± 0.421


Q3Y_S12L_S60A_L79Y
1.993 ± 0.485


Q3Y_S7Q_S60A_L79Y
1.461 ± 0.475


Q3Y_S7Q_S60A_F83Y
1.982 ± 0.069


Q3Y_S12L_S60A_F83Y
1.035 ± 0.312


Q3Y_S7Q_S12L_F83Y
1.729 ± 0.186


Q3Y_S60A_L79Y_F83Y
1.966 ± 0.372


Q3Y_S7Q_S12L_S60A
1.037 ± 0.089


Q3Y_S12L_L79Y_F83Y
1.298 ± 0.513


Q3Y_S7Q_L79Y_F83Y
2.133 ± 0.038


S7Q_S12L_L79Y_F83Y
1.397 ± 0.487


S7Q_S12L_S60A_F83Y
1.281 ± 0.406


S7Q_S12L_S60A_L79Y
4.182 ± 1.036


S7Q_S60A_L79Y_F83Y
5.438 ± 2.931


Q3Y_S7Q_S12L_L79Y
1.096 ± 0.095


S7Q_S12L_L79Y_F83Y
2.795 ± 0.188


S12L_S60A_L79Y_F83Y
0.721 ± 0.235


Q3Y_S7Q_S12L_S60A_L79Y_F83Y
1.421 ± 0.245









Example 3
Testing Variants Incorporating Other Amino Acids with Same Side-Chain Functionalities

In order to ascertain whether the amino acid substitutions are representative of the other amino acids that share the same side-chain functionalities, the following additional TNFα dAb variants were tested for their TNFα neutralization potency:


(1) Q3F


(2) S7N, S7G


(3) S12V, S12T


(4) S60E, S60G


(5) L79F, L79G


These substitutions were selected as they belong to the same amino acid functional class as those identified as improved dAb variants. For example, Q3Y exhibited improved potency, therefore in this Example, Q3F was tested to determine whether the substitution of Q3 into another amino acid containing an aromatic side chain would improve potency. Variants that contain substitutions that belong to the same amino acid class as the amino acid in the original unmodified dAb construct were also tested. These are listed above as the second substitution. For example, as S7Q variant was identified as one with improved potency, S7N was tested here as Q and N are amino acids that have amide in their side-chains and further, S7G was also tested as G is an amino acid with a small side chain, like S in the unmodified construct.


These variants were produced and tested as described above. Briefly, gene fragments encoding the selected substitutions were synthesized and inserted into a vector suitable for mammalian cell expression. The variants were transiently expressed in suspension variant of the CHOK1 cell line using the Freestyle™ MAX CHO transfection method. The conditioned media was subjected to Protein A affinity chromatography and the purified protein quantitated by BCA® assay. These variants were assayed for their ability to neutralize TNFα-mediated cytotoxicity using the standard format of the L929 neutralization assay described in Example 2.


The EC50 values of variants tested in this Example are also listed in Table 3, together with data of the combination variants tested also using the standard 11-point format of the L929 assay.


This Example showed that the use of the nine amino acids as representatives of the different amino acid classes was justified (see Example 1 for the categories that the amino acids represent). All tested variants comprising substitutions into another amino acid of the same amino acid class displayed improved potency as compared to the initially identified variants, except for L79F and S60E that exhibited similar TNFα neutralization potency.


Example 4
Variants of Ferritin Targeted Domain Antibodies

This Example set out to test whether framework variants that resulted in improved function of the TNFα dAb can be applied to domain antibodies of different specificities.


A different domain antibody was selected—a light chain variable VL dAb targeted to human ferritin. The sequence of the anti-ferritin dAb is SEQ ID NO. 4.


In the process of designing these dAb variants, the dAb protein sequences were aligned using ClustalW (Higgins et al., 1994). Three dimensional models of VL dAbs were constructed and examined using the MODELER program (Sali and Blundell, 1993; Accelrys® Discovery Studio® v2.0.1.7347). Six framework variants that resulted in improved function of the TNFα dAb selected for this Example were position 3 to Y, position 7 to Q, 12 to L, 60 to A, 79 to Y and 83 to Y.


Ferritin VL dAb was very similar in sequence and structure, in particular in the framework region, when compared to the TNFα VL dAb. The six selected framework residues were located at similar spatial positions in both the VL dAbs, and therefore, six variants comprising of single substitution mutations at these framework regions of the ferritin dAb were made. Like the TNFα dAb variants, the ferritin VL dAb variants were also produced as Fc fusion constructs. The sequence of the unmodified ferritin VL dAb Fc construct is set out in SEQ ID NO 5. The variants produced were Q3Y, S7Q, S12L, S60A, Q79Y and F83Y and are herein collectively referred to as Territin dAb Fc variants'.


The gene fragments encoding the variants were optimized for expression in mammalian cells. They were assembled from synthetic oligonucleotides and/or PCR products and cloned into a vector suitable for mammalian cell expression. The final DNA constructs were verified by sequencing.


The Ferritin dAb Fc framework variants were transiently expressed in suspension variant of the CHOK1 cell line. Transfections were performed using Freestyle™ MAX CHO transfection method. Six days after transfection conditioned media was harvested from by centrifugation and the supernatants filtered using 0.22 μm membrane filter. The variants were purified by Protein A affinity chromatography. After sample loading, unbound proteins were washed off using PBS, variants were eluted using 0.1 M citric acid pH 3.5 and fractions neutralized with the addition of 1 M Tris pH 9.0 then concentrated and buffer exchanged into PBS using sample concentrators (namely Micron YM-30 units or Amicon® Ultra-4 devices; Millipore®). Protein concentration was determined by BCA® assay.


Testing of Ferritin VL dAb Variants

The six Ferritin dAb Fc framework variants (Q3Y, S7Q, S12L, S60A, Q79Y and F83Y) as well as unmodified Ferritin dAb Fc construct were assessed for their ability to bind target antigen, human spleen ferritin, in a ferritin binding ELISA.


Ferritin Binding ELISA

Ninety-six well flat-bottom plates (MaxiSorp™, Nunc™) were coated with Ferritin dAb Fc proteins (10 μg/ml) in carbonate coating buffer overnight at 4° C. After the overnight incubation, the plates were washed three times in PBS-T then PBS followed by incubation in blocking solution (1% w/v BSA diluted in PBS) for 1 hour at room temperature. After washing, human spleen ferritin (AppliChem) serially diluted 1 in 2 from 126 μg/ml to 0.246 μg/ml in diluent buffer was added to the wells and incubated for 2 hours at room temperature. The plates were washed in PBS-T and PBS then incubated with secondary antibody HRP ferritin polyclonal (Abcam®; 1:2000 in diluent buffer). After one hour incubation at room temperature, plates were washed as above then 100 μl TMB solution added to the wells. Colorimetric development was performed at room temperature and stopped with 50 μl 1 M hydrochloric acid and absorbance read at 450 nm.



FIG. 3 illustrates the ability of the Ferritin dAb Fc proteins to bind human spleen ferritin, as assayed in the ferritin binding ELISA.


All Ferritin dAb Fc variants comprising the single framework substitutions were able to bind more ferritin than the unmodified Ferritin dAb Fc protein.


BIBLIOGRAPHY



  • Ashkenazi et al. (1990). Mapping the CD4 binding site for human immunodeficiency virus by alanine-scanning mutagenesis. Proc Natl Acad Sci USA 87: 7150-7154

  • Barbas et al. (1992). Semisynthetic combinatorial antibody libraries: a chemical solution to the diversity problem. Proc Natl Acad Sci USA 89: 4457-4461

  • Barbas et al. (1992). Human monoclonal Fab fragments derived from a combinatorial library bind to respiratory syncytial virus F glycoprotein and neutralize infectivity. Proc Natl Acad Sci USA 89: 10164-10168.

  • Barderas et al. (2008). Affinity maturation of antibodies assisted by in silico modelling. Proc Natl Acad Sci USA 105: 9029-9034

  • Berzofsky et al. (1984). Antibody-Antigen Interactions. In Fundamental Immunology. Paul (Ed.) Raven Press: New York

  • Breitling et al. (1991). A surface expression vector for antibody screening. Gene 104: 147-153

  • Boder et al. (2000). Directed evolution of antibody fragments with monovalent femtomolar antigen-binding affinity. Proc Natl Acad Sci USA 97: 18870-18877

  • Burton et al. (1991). A large array of human monoclonal antibodies to type 1 human immunodeficiency virus from combinatorial libraries of asymptomatic seropositive individuals. Proc Natl Acad Sci USA 88:10134-10137

  • Cadwell and Joyce (1992). Randomization of genes by PCR mutagenesis. PCR Methods Appl. 2: 28-23

  • Caton and Koprowski (1990). Influenza virus hemagglutinin-specific antibodies isolated from a combinatorial expression library are closely related to the immune response of the donor. Proc Natl Acad Sci USA 87:6450-6454

  • Chang et al. (1991). Expression of antibody Fab domains on bacteriophage surfaces. Potential use for antibody selection. Journal of Immunology 147: 3610-3614

  • Chatellier et al. (1995). Codon-based combinatorial alanine scanning site-directed mutagenesis: design, implementation, and polymerase chain reaction screening. Analytical Biochemistry 229: 282-290

  • Chen et al. (1999). Selection and analysis of an optimized anti-VEGF antibody: crystal structure an affinity matured Fab in complex with antigen. Journal of Molecular Biology 293: 865-881

  • Clackson et al. (1991). Making antibody fragments using phage display libraries. Nature 352: 624-628

  • Chothia et al. (1989). Conformations of immunoglobulin hypervariable regions. Nature 342: 877-883

  • Chothia and Lesk (1987). Canonical structures for the hypervariable regions of immunoglobulins. Journal of Molecular Biology 196: 901-917

  • Chen et al. (1999). Selection and analysis of an optimized anti-VEGF antibody: crystal structure of an affinity-matured fab in complex with antigen. Journal of Molecular Biology 293: 865-881

  • Coia et al. (2001). Protein affinity maturation involving E. coli mutator cells. Journal of Immunological Methods 251: 181-193

  • Cunnighman and Wells (1989). High-resolution epitope mapping of hGH-receptor interactions by alanine-scanning mutagenesis. Science 244: 1081-1085

  • Daugherty et al. (1998). Antibody affinity maturation using bacterial surface display. Protein Engineering 11: 825-832

  • de Kruif et al. (1995). Rapid selection of cell subpopulation-specific human monoclonal antibodies from a synthetic phage antibody library. Proc Natl Acad Sci USA 92: 3938-3942

  • Dooley and Flajnik (2005). Shark immunity bites back: affinity maturation and memory response in the nurse shark, Ginglymostoma cirratum. European Journal of Immunology 35:936-945.

  • Ellington and Szostak (1990). In vitro selection of RNA molecules that binding specific ligands. Nature 346: 818-822

  • Griffiths et al. (1994). Isolation of high affinity human antibodies directly from large synthetic repertoires. EMBO J. 13: 3245-3260

  • Finlay et al. (2009). Affinity maturation of a humanized rat antibody for anti-RAGE therapy: comprehensive mutagenesis reveals a high level of mutational plasticity both inside and outside the complementarity-determining regions. Journal of Molecular Biology 388: 541-558

  • Fischmann et al. (1991). Crystallographic refinement of the three-dimensional structure of the FabD1.3-lysozyme complex at 2.5-A resolution. Journal of Biological Chemistry 266: 12915-12920

  • Foote and Winter (1992). Antibody framework residues affecting the conformation of the hypervariable loops. Journal of Molecular Biology 224: 487-499

  • Fromant et al. (1995). Direct random mutagenesis of gene sized DNA fragments using polymerase chain reaction. Analytical Biochemistry 224: 347-353

  • Hamers-Casterman et al. (1993). Naturally occurring antibodies devoid of light chains. Nature 363: 446-448

  • Harayama (1998). Artificial evolution by DNA shuffling. Trends in Biotechnology 16: 258-261

  • Hawkins et al. (1992). Selection of phage antibodies by binding affinity. Mimicking affinity maturation. Journal of Molecular Biology 226: 889-896

  • Hawkins and Winter (1992). Cell selection strategies for making antibodies from variable gene libraries: trapping the memory pool. European Journal of Immunology 22: 867-870

  • Higgins et al. (1994). CLUSTAL W: improving the sensitivity of progressive multiple sequence alignment through sequence weighting, position-specific gap penalties and weight matrix choice. Nucleic Acids Research 22: 4673-4680

  • Ho et al. (2005). In vitro antibody evolution targeting germline hot spots to increase activity of an anti-CD22 immunotoxin. Journal of Biological Chemistry 280: 607-617

  • Holliger et al. (1993). “Diabodies”: small bivalent and bispecific antibody fragments. Proc Natl Acad Sci USA 90: 6444-6448

  • Holt et al. (2003). Domain antibodies: proteins for therapy. Trends in Biotechnology 21: 484-490

  • Honegger and Pluckthun (2001). Yet another numbering scheme for immunoglobulin variable domains: an automatic modelling and analysis tool. Journal of Molecular Biology 309: 657-670

  • Hoogenboom and Winter (1992). By-passing immunisation. Human antibodies from synthetic repertoires of germline VH gene segments rearranged in vitro. Journal of Molecular Biology 227: 381-388

  • Hoogenboom et al. (1991). Multi-subunit proteins on the surface of filamentous phage: methodologies for displaying antibody (Fab) heavy and light chains. Nucleic Acids Research 19: 4133-4137

  • Huse et al. (1989). Generation of a large combinatorial library of the immunoglobulin repertoire in phage lambda. Science 246: 1275-1281

  • Irving et al. (1996). Affinity maturation of recombinant antibodies using E. coli mutator cells. Immunotechnology 2: 127-143

  • Jackson et al. (1995). In vitro antibody maturation. Journal of Immunology 154: 3310-3319

  • Johnson and Wu (2001) Kabat Database and its applications: future directions. Nucleic Acids Res. 29: 205-206.

  • Junginger et al. (1994). In Drug Permeation Enhancement. Hsieh (Eds.) Marcel Dekker Inc: New York 59-90

  • Kabat et al. (1983). Sequence of Proteins of Immunological Interest. US Department of Health and Human Services.

  • Kang et al. (1991a). Linkage of recognition and replication functions by assembling combinatorial antibody Fab libraries along phage surfaces. Proc Natl Acad Sci USA 88: 4363-4366

  • Kang et al. (1991b). Antibody redesign by chain shuffling from random combinatorial immunoglobulin libraries. Proc Natl Aca Sci USA. 88: 11120-11123

  • Kast and Hilvert (1997). 3D structural information as a guide to protein engineering using genetic selection. Current Opinion in Structural Biology 7: 470-479

  • Kettleborough et al. (1991). Humanization of a mouse monoclonal antibody by CDR-grafting: the importance of framework residues on loop conformation. Protein Engineering 4: 773-783

  • Knappick et al. (2000). Fully synthetic human combinatorial antibody libraries (HuCAL) based on modular consensus frameworks and CDRs randomized with trinucleotides. Journal of Molecular Biology 296: 57-86

  • Koide et al. (2007). Exploring the capacity of minimalist protein interfaces: interface energetics and affinity maturation to picomolar KD of a single-domain antibody with a flat paratope. Journal of Molecular Biology 373: 941-953

  • Kuby (1992). Immunology. Freeman and Company: New York

  • Jones (1848). On a new substance occurring in the urine of a patient with “mollities ossium.” Phil. Trans Roy Soc London 55-62

  • Jones et al. (1986). Replacing the complementarity-determining regions in a human antibody with those from a mouse. Nature 321: 522-525

  • Lee et al. (2004). High-affinity human antibodies from phage-displayed synthetic Fab libraries with a single framework scaffold. Journal of Molecular Biology 340: 1073-1093

  • LeFranc (2003). IMGT, the international ImMunoGeneTics Database®. Nucleic Acids Research. Nucleic acids Research 31: 307-310

  • Low et al. (1996). Mimicking somatic hypermutation: affinity maturation of antibodies displayed on bacteriophage using a bacterial mutator strain. Journal of Molecular Biology 260: 359-368

  • Lowman et al. (1991). Selecting high-affinity binding proteins by monovalent phage display. Biochemistry 30: 10832-10838.

  • Marks et al. (1991). By-passing immunization. Human antibodies from V-gene libraries displayed on phage. Journal of Molecular Biology 222: 581-597

  • Marks et al. (1992). Molecular evolution of proteins on filamentous phage. Mimicking the strategy of the immune system. Journal of Biological Chemistry 267: 16007-160010

  • Marks (2004). Antibody affinity maturation by chain shuffling. Methods in Molecular Biology 248: 327-343

  • McCafferty et al. (1991). Phage-enzymes: expression and affinity chromatography of functional alkaline phosphatase on the surface of bacteriophage. Protein Engineering 4: 955-961

  • McCafferty et al. (1990). Phage antibodies: filamentous phage displaying antibody variable domains. Nature 348: 552-554

  • Mullinax et al. (1990). Identification of human antibody fragment clones specific for tetanus toxoid in a bacteriophage lambda immunoexpression library. Proc Natl Acad Sci USA 87: 8095-8099

  • Murase et al. (2003). EF-Tu binding peptides identified, dissected, and affinity optimized by phage display. Chemistry and Biology 10: 161-168

  • Muyldermans et al. (1994). Sequence and structure of VH domain from naturally occurring camel heavy chain immunoglobulins lacking light chains. Protein Engineering 7:1129-1135

  • Nissim et al. (1994). Antibody fragments from a ‘single pot’ phage display library as immunochemical reagents. EMBO J. 13: 692-698

  • Pal et al. (2005). Alternative views of functional protein binding epitopes obtained by combinatorial shotgun scanning mutagenesis. Protein Science 14: 2405-2413

  • Persson et al. (1991). Generation of diverse high-affinity human monoclonal antibodies by repertoire cloning. Proc Natl Acad Sci USA 88: 2432-2436

  • Qin et al. (2007). A novel domain antibody rationally designed against TNFα using variable region of human heavy chain antibody as scaffolds to display antagonistic peptides. Molecular Immunology 44: 2355-2361

  • Queen et al. (1989). A Humanized Antibody That Binds To The Interleukin 2 Receptor. Proc Natl Acad Sci USA 86: 10029-10033

  • Retter et al. (2005). VBASE2, an integrative V gene database. Nucleic Acids Res. 33: D671-4

  • Riechmann et al. (1988). Reshaping Human Antibodies For Therapy. Nature 332: 323-327

  • Ridgway et al. (1999). Identification of a Human anti-CD55 single-chain Fv by subtractive panning of a phage library using tumour and non tumour cell lines. Cancer Research 59: 2718-2723

  • Robinson (Ed.) (1994). Sustained and controlled release drug delivery systems. Marcel Dekker Inc: NY

  • Sali and Blundell (1993). Comparative protein modelling by satisfaction of spatial restraints. Journal of Molecular Biology 234: 779-815

  • Schier et al. (1996). Isolation of picomolar affinity anti-c-erbB-2 single-chain Fv by molecular evolution of the complementarity determining regions in the center of the antibody binding site. Journal of Molecular Biology 263: 551-567

  • Scott and Smith (1990). Searching for peptide ligands with an epitope library. Science 249: 386-390

  • Stemmer (1994). Rapid evolution of a protein in vitro by DNA shuffling. Nature 370: 389-391

  • Streltsov et al. (2004). Structural evidence for evolution of shark Ig new antigen receptor variable domain antibodies from a cell-surface receptor. Proc Natl Acad Sci USA 101:12444-12449.

  • Tawfik and Griffths (1998). Man-made cell-like compartments for molecular evolution. Nature Biotechnology 16: 652-656

  • Thom et al. (2006). Probing a protein-protein interaction by in vitro evolution. Proc Natl Acad Sci USA 103: 7619-7624

  • Thompson et al. (1996). Affinity maturation of a high-affinity human monoclonal antibody against the third hypervariable loop of human immunodeficiency virus: use of phage display to improve affinity and broaden strain reactivity. Journal of Molecular Biology 256: 77-88

  • Tuerk and Gold (1990). Systematic evolution of ligands by exponential enrichment: RNA ligands to bacteriophage T4 DNA polymerase. Science 249: 505-510

  • Tulip et al. (1992). Refined crystal structure of the influenza virus N9 neuraminidase-NC41 Fab complex. Journal of Molecular Biology 227: 122-148

  • Vajdos et al. (2002). Comprehensive functional maps of the antigen-binding site of an anti-ErbB2 antibody obtained with shotgun scanning mutagenesis. Journal of Molecular Biology 320: 415-428

  • Valjakka et al. (2002). Crystal structure of an in vitro affinity- and specificity-matured anti-testosterone Fab in complex with testosterone. Improved affinity results from small structural changes within the variable domains. Journal of Biological Chemistry 277: 44021-44027

  • van den Beucken et al. (2003). Affinity maturation of Fab antibody fragments by fluorescent-activated cell sorting of yeast-displayed libraries. FEBS letters 546: 288-294

  • Vaughan et al. (1996). Human antibodies with sub-nanomolar affinities isolated from a large non-immunized phage display library. Nature Biotechnology 14: 309-314

  • Verhoeyen et al. (1988). Reshaping human antibodies: grafting an antilysozyme activity. Science 239: 1534-1536

  • Ward et al. (1989). Binding activities of a repertoire of single immunoglobulin variable domains secreted from Escherichia coli. Nature 341: 544-546

  • Weiss et al. (2000). Rapid mapping of functional protein epitopes by combinatorial alanine-scanning. Proc Natl Acad Sci USA 97: 8950-8954

  • Wu et al. (1998). Stepwise in vitro affinity maturation of Vitaxin, an alphav beta3-specific humanized mAb. Proc. Natl. Acad. Sci. U.S.A. 95: 6037-6042

  • Xiang et al. (1999). Light-chain framework region residue Try71 of chimeric B72.3 antibody plays an important role in influencing TAG72 antigen binding. Protein Engineering 12: 417-421

  • Yang et al. (1995). CDR walking mutagenesis for the affinity maturation of a potent human anti-HIV-1 antibody into the picomolar range. Journal of Molecular Biology 254: 392-403

  • Yau et al. (2005). Affinity maturation of a V(H)H by mutational hotspot randomization. Journal of Immunological Methods 297: 213-224

  • Yelton et al. (1995). Affinity maturation of the BR96 anti-carcinoma antibody by codon-based mutagenesis. Journal of Immunology 155: 1994-2004

  • Yoon et al. (2006). Construction, affinity maturation, and biological characterization of an anti-tumour-associated glycoprotein-72 humanized antibody. Journal of Biological Chemistry 281: 6985-6992

  • Zha et al. (2003). Family shuffling with single-stranded DNA. Methods in Molecular Biology 182: 243-257

  • Zahnd et al. (2004). Directed in vitro evolution and crystallographic analysis of a peptide-binding single chain antibody fragment (scFv) with low picomolar affinity. Journal of Biological Chemistry 279: 18870-18877

  • Zhang et al. (2004). Improved breadth and potency of a HIV-1 neutralizing human single-chain antibody by random mutagenesis and sequence antigen panning. Journal of Molecular Biology 335: 209-219


Claims
  • 1-33. (canceled)
  • 34. A modified domain antibody in which the framework sequences of the unmodified domain antibody are as set out in SEQ ID NO 1 and wherein the modified domain antibody includes at least one substitution selected from the group consisting of D1A, D1Y, D1K, I2L, I2P, Q3Y, Q3F, Q3D, Q3H, S7A, S7G, S7D, S7Q, S7N, S9A, S10A, S10L, L11Q, S12A, S12L, S12V, S12T, S12Q, S14A, D17Y, D17H, R18D, R185, T20Y, T22A, T22Y, T22Q, T22H, T22K, Q38H, P40D, K42A, K425, K42H, P44L, K45S, K45L, K45Y, K45D, K45H, 148A, Y49S, Y49D, Y49H, G57A, G57S, G57L, G57Y, G57Q, G57H, G57K, P59A, P59S, P59D, P59H, S60A, S60G, S60L, S60Y, S60D, S60Q, S60H, S60P, S60K, R61H, F62Y, T69D, D70Q, L79A, L79G, L79S, L79Y, L79V, L79D, L79H, L79K, F83A, F83S, F83L, F83Y, F83D and conservative substitutions thereof and combinations thereof.
  • 35. A modified domain antibody as claimed in claim 34 which comprises a substitution selected from the group consisting of L79G, L79S, L79Y, L79K, L79D, L79A, L79V and L79H.
  • 36. A modified domain antibody as claimed in claim 34 which comprises a substitution selected from the group consisting of S60G, S60A, S60D, S60L, S60H, S60Q, S60Y, S60P and S60K.
  • 37. A modified domain antibody as claimed in claim 34 which comprises a substitution selected from the group consisting of S12T, S12L, S12V, S12A and S12Q.
  • 38. A modified domain antibody as claimed in claim 34 which comprises a substitution selected from the group consisting of F83Y, F83L, F83A, F83S and F83D.
  • 39. A modified domain antibody as claimed in claim 34 which comprises a substitution selected from the group consisting of P59A, P59S, P59D and P59H.
  • 40. A modified domain antibody as claimed in claim 34 which comprises a substitution selected from the group consisting of G57S, G57Q, G57H, G57A, G57L, G57Y and G57K.
  • 41. A modified domain antibody as claimed in claim 34 which comprises a substitution selected from the group consisting of Q3F, Q3Y, Q3H and Q3D.
  • 42. A modified domain antibody as claimed in claim 34 which comprises a substitution selected from the group consisting of S7A, S7N, S7G, S7Q and S7D.
  • 43. A domain antibody as claimed in claim 34 in which the domain antibody is attached to an Fc or modified Fc.
  • 44. A domain antibody as claimed in claim 43 in which the sequence of the modified Fc is SEQ ID NO 6 or SEQ ID NO 7.
  • 45. A nucleic acid molecule encoding the domain antibody of claim 34.
  • 46. A nucleic acid molecule as claimed in claim 45 in which the nucleic acid molecule has a sequence selected from SEQ ID NO. 8 to SEQ ID NO. 708.
  • 47. A transformed cell comprising the nucleic acid molecule of claim 45 wherein the cell expresses the nucleic acid molecule.
  • 48. A domain antibody which binds TNFα, the domain antibody having SEQ ID NO 1 wherein at least one amino acid in the framework regions is substituted, wherein the domain antibody has improved TNFα neutralising potency in comparison to the domain antibody of SEQ ID NO 1.
  • 49. A domain antibody as claimed in claim 48 in which the substitution is selected from the group consisting of D1A, D1Y, D1K, I2L, I2P, Q3F, Q3Y, Q3D, Q3H, S7A, S7D, S7Q, S7G, S7N, S9A, S10A, S10L, S12A, S12L, S12Q, S12V, S12T, S14A, R18D, T22A, K42A, K45S, Y49H, G57S, G57Q, G57H, P59A, P59S, P59D, S60G, S60A, S60L, S60D, S60Q, S60H, F62Y, L79G, L79S, L79Y, L79D, L79K, F83A, F83L, F83S, F83Y and conservative substitutions thereof and combinations thereof.
  • 50. A domain antibody as claimed in claim 49 which comprises a substitution selected from the group consisting of L79G, L79S, L79Y, L79K and L79D.
  • 51. A domain antibody as claimed in claim 49 which comprises a substitution selected from the group consisting of S60G, S60A, S60D, S60L, S60H and S60Q.
  • 52. A domain antibody as claimed in claim 49 which comprises a substitution selected from the group consisting of S12T, S12L, S12V, S12A and S12Q.
  • 53. A domain antibody as claimed in claim 49 which comprises a substitution selected from the group consisting of F83Y, F83L, F83A and F83S.
  • 54. A domain antibody as claimed in claim 49 which comprises a substitution selected from the group consisting of P59A, P59S and P59D.
  • 55. A domain antibody as claimed in claim 49 which comprises a substitution selected from the group consisting of G57S, G57Q and G57H.
  • 56. A domain antibody as claimed in claim 49 which comprises a substitution selected from the group consisting of Q3F, Q3Y, Q3H and Q3D.
  • 57. A domain antibody as claimed in claim 49 which comprises a substitution selected from the group consisting of S7A, S7N, S7G, S7Q and S7D.
  • 58. A domain antibody as claimed in claim 48 in which the domain antibody includes multiple substitutions.
  • 59. A domain antibody as claimed in claim 58 in which the multiple substitutions are selected from the group consisting of S60A_F83Y, Q3Y_S12L, S12L_S60A_L79Y_F83Y, S7Q_S60A, S7Q_S12L, Q3Y_S7Q, S12L_L79Y_F83Y, Q3Y_S60A, S12L_S60A_F83Y, S7Q_L79Y_F83Y, Q3Y_S12L_S60A_F83Y, Q3Y_S7Q_S12L_S60A, Q3Y_S60A_F83Y, Q3Y_S7Q_S12L, Q3Y_S7Q_S12L_L79Y, Q3Y_S12L_L79Y, Q3Y_L79Y, S7Q_S12L_S60A_F83Y, L79Y_F83Y, Q3Y_S12L_L79Y_F83Y, S60A_L79Y_F83Y, S7Q_S12L_L79Y_F83Y, Q3Y_S7Q_S12L_S60A_L79Y_F83Y, Q3Y_S7Q_S60A, Q3Y_S7Q_S60A_L79Y and S12L_S60A_L79Y.
  • 60. A domain antibody as claimed in claim 48 in which the domain antibody is attached to an Fc or modified Fc.
  • 61. A domain antibody as claimed in claim 60 in which the sequence of the modified Fc is SEQ ID NO 6 or SEQ ID NO 7.
  • 62. A nucleic acid molecule encoding the domain antibody of claim 48.
  • 63. A nucleic acid molecule as claimed in claim 62 in which the nucleic acid molecule has a sequence selected from SEQ ID NO. 8 to SEQ ID NO. 708.
  • 64. A transformed cell comprising the nucleic acid molecule of claim 62 wherein the cell expresses the nucleic acid molecule.
  • 65. A domain antibody which binds TNFα, the domain antibody having SEQ ID NO 1 wherein at least one amino acid in the framework regions is substituted, wherein the domain antibody has improved TNFα binding and/or improved protein expression levels in comparison to the domain antibody of SEQ ID NO 1.
  • 66. A domain antibody as claimed in claim 65 in which the substitution is selected from the group consisting of S7A, L11Q, D17Y, D17H, R18S, T20Y, T22Y, T22Q, T22H, T22K, Q38H, P40D, K425, K42H, P44L, K45L, K45Y, K45D, K45H, 148A, Y49S, Y49D, Y49H, G57A, G57S, G57L, G57Y, G57Q, G57H, G57K, P59S, P59D, P59H, S60A, S60L, S60Y, S60D, S60Q, S60H, S60P, S60K, R61H, T69D, D70Q, L79A, L79S, L79Y, L79D, L79H, L79K, F83A, F83Y, F83D and conservative substitutions thereof and combinations thereof.
  • 67. A domain antibody as claimed in claim 65 in which the substitution is selected from the group consisting of S7A, Y49H, G57S, G57Q, G57H, P59S, P59D, S60A, S60L, S60D, S60Q, S60H, L79S, L79Y, L79D, L79K, F83A, F83Y and conservative substitutions thereof and combinations thereof.
  • 68. A domain antibody as claimed in claim 65 in which the domain antibody is attached to an Fc or modified Fc.
  • 69. A domain antibody as claimed in claim 68 in which the sequence of the modified Fc is SEQ ID NO 6 or SEQ ID NO 7.
  • 70. A nucleic acid molecule encoding the domain antibody of claim 65.
  • 71. A nucleic acid molecule as claimed in claim 70 in which the nucleic acid molecule has a sequence selected from SEQ ID NO. 8 to SEQ ID NO. 708.
  • 72. A transformed cell comprising the nucleic acid molecule of claim 70 wherein the cell expresses the nucleic acid molecule.
  • 73. A VL domain antibody comprising a VL framework acceptor sequence wherein the framework sequence has been modified such that the framework comprises at least one amino acid substitution at least one position selected from the group consisting of 3, 7, 12, 60, 79, 83 and combinations thereof.
  • 74. A VL domain antibody as claimed in claim 73 wherein the domain antibody comprises at least one framework substitution selected from the group consisting of Y at position 3, Q at position 7, L at position 12, A at position 60, Y at position 79, Y at position 83 and combinations thereof.
  • 75. A domain antibody as claimed in claim 73 in which the domain antibody is attached to an Fc or modified Fc.
  • 76. A domain antibody as claimed in claim 75 in which the sequence of the modified Fc is SEQ ID NO 6 or SEQ ID NO 7.
Priority Claims (2)
Number Date Country Kind
2008904175 Aug 2008 AU national
2009902142 May 2009 AU national
PCT Information
Filing Document Filing Date Country Kind 371c Date
PCT/AU2009/001043 8/13/2009 WO 00 5/11/2011
Provisional Applications (1)
Number Date Country
61089019 Aug 2008 US