OA16601A - Bispecific molecules binging to VEGF and Ang2. - Google Patents

Bispecific molecules binging to VEGF and Ang2. Download PDF

Info

Publication number
OA16601A
OA16601A OA1201300405 OA16601A OA 16601 A OA16601 A OA 16601A OA 1201300405 OA1201300405 OA 1201300405 OA 16601 A OA16601 A OA 16601A
Authority
OA
OAPI
Prior art keywords
amino acid
seq
binding
vegf
vhhs
Prior art date
Application number
OA1201300405
Inventor
Andreas Gschwind
Rene Georg Ott
Joachim Boucneau
Marie-Ange Buyse
Erik Depla
Original Assignee
Boehringer Ingelheim International Gmbh
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Boehringer Ingelheim International Gmbh filed Critical Boehringer Ingelheim International Gmbh
Publication of OA16601A publication Critical patent/OA16601A/en

Links

Abstract

Bispecifc binding molecules binding to both VEGF and Ang2, preferably in the form of immunoglobulin single variable domains like VHHs and domain antibodies, pharmaceutical compositions containing the same and their use in the treatment of diseases that are associated with VEGF- and/or Ang2- mediated effects on angiogenesis are disclosed. Further, nucleic acids encoding bispecific binding molecules, host cells and methods for preparing same are also described.

Description

The invention relates to the field of human therapy, in particular cancer therapy and agents and compositions useful in such therapy.
BACKGROUND OF THE INVENTION
When tumors reach a critical size of approximately 1 mm3 they become dépendent on angiogenesis for maintaining blood supply with oxygen and nutritients to allow for further growth. Anti-angiogenesis thérapies hâve become an important treatment option for several types of tumors. These thérapies hâve focused on blocking the VEGF pathway (Ferrara et al., Nat Rev Drug Discov. 2004 May;3(5):391-400.) by neutralizing VEGF (Avastin) or its receptors (Sutent and Sorafinib). Recent studies in mice hâve shown, that Angiopoietin2 (Ang2), a ligand of the Tie2 receptor, controls vascular remodeling by enabling the functions of other angiogenic factors, such as VEGF. Ang2 is primarily expressed by endothélial cells, strongly induced by hypoxia and other angiogenic factors and has been demonstrated to regulate tumor vessel plasticity, allowing vessels to respond to VEGF and FGF2 (Augustin et al., Nat Rev Mol Cell Biol. 2009 Mar; 10(3):165-77.). Consistent with this rôle, the délétion or inhibition of Ang2 results in reduced angiogenesis (Gale et al., Dev Cell. 2002 Sep;3(3):302-4.) (Falcôn et al., Am J Pathol. 2009 Nov;175(5):2159-70.). Elevated Ang2 sérum concentrations hâve been reported for patients with colorectal cancer, NSCLC and melanoma (Goede et al., Br J Cancer. 2010 Oct 26;103(9):1407-14),(Park et al., Chest. 2007 Jul;132(1): 200-6.),(Helfrich étal., Clin Cancer Res. 2009 Feb 15;15(4):1384-92.). In CRC cancer Ang2 sérum levels correlate with therapeutic response to anti-VEGF therapy.
The Ang-Tie System consists of 2 receptors (Tie1 and Tie2) and 3 ligands (Ang1, Ang2 and Ang4) (Augustin et al., Nat Rev Mol Cell Biol. 2009 Mar;10(3):165-77.).
Tie2, Ang1 and Ang2 are the best studied members of this family, Tie1 is an orphan receptor and the rôle of Ang4 for vascular remodelling still needs to be defined. Ang2 and Ang1 médiate opposing functions upon Tie2 binding and activation. Ang2mediated Tie2 activation results in endothélial cell activation, pericyte dissociation, vessel leakage and induction of vessel sprouting. In contrast to Ang2, Ang1 signaling maintains vessel integrity by recruitment of pericytes, thereby maintaining endothélial cell quiescence.
Angiopoietin 2 (Ang2) is a secreted, 66 kDa ligand for the Tie2 receptor tyrosine kinase (Augustin et al., Nat Rev Mol Cell Biol. 2009 Mar;10(3):165-77.). Ang2 consists of an N-terminal coiled-coil domain and a C-terminal fibrinogen-like domain, the latter is required for Tie2 interaction. Ang2 is primarily expressed by endothélial cells and strongly induced by hypoxia and other angiogenic factors, including VEGF. Tie2 is found on endothélial cells, haematopoietic stem cells and tumor cells. Ang2Tie2 has been demonstrated to regulate tumor vessel plasticity, allowing vessels to respond to VEGF and FGF2,
In vitro Ang2 has been shown to act as a modest mitogen, chemoattractant and inducer of tube formation in human umbilical vein endothélial cells (HUVEC). Ang2 induces tyrosine phosphorylation of ectopically expressed Tie2 in fibroblasts and promûtes downstream signaling events, such as phosphorylation of ERK-MAPK, AKT and FAK in HUVEC. An antagonistic rôle of Ang2 in Ang1-induced endothélial cell responses has been described.
Ang2 -deficiency has been shown to resuit in a profound lymphatic patterning defect in mice. Although the loss of Ang2 is dispensable for embryonic vascular development, Ang2 -déficient mice hâve persistent vascular defects in the retina and kidney. Together with the dynamic pattern of Ang2 expression at sites of angiogenesis (for example ovary), these findings indicate that Ang2 controls vascular remodeling by enabling the functions of other angiogenic factors, such as VEGF.
The Ang2-Tie2 System exerts crucial rôles during the angiogenic switch and later stages of tumor angiogenesis. Ang2 expression is strongly up-regulated in the tumor associated endothélium, Reduced growth of tumors has been observed when implanted into Ang2 -déficient mice, especially during early stages of tumor growth. Therapeutic blocking of Ang2 with Ang2 mAbs has shown broad efficacy in a variety of tumor xenograft models. Additive effects of Ang2 mAbs with inhibitors of VEGFR2 (mAbs and small molecular weight inhibitors) hâve been described.
As described in e.g. US2008/0014196 and W02008/101985, angiogenesis is implicated in the pathogenesis of a number of disorders, including solid tumors and metastasis as well as eye diseases. One of the most important pro-angiogenic factors is vascular endothélial growth factor (VEGF), also termed VEGF-A or vascular permeability factor (VPF), VEGF belongs to a gene family that includes placenta growth factor (PIGF), VEGF-B, VEGF-C, VEGF-D, VEGF-E and VEGF-F. Alternative splicing of mRNA of a single gene of human VEGF results in at least six isoforms (VEGF121, VEGF145, VEGF165, VEGF183, VEGF189, and VEGF206), VEGF165 being the most abundant isoform.
Two VEGF tyrosine kinase receptors (VEGFR) hâve been identified that interact with VEGF, i.e. VEGFR-1 (also known as Flt-1) and VEGFR-2 (also known as KDR or FIK-1 ). VEGFR-1 has the highest affinity for VEGF, while VEGFR- 2 has a somewhat lower affinity for VEGF. Ferrara (Endocrine Rev. 2004, 25: 581-611) provide a detailed description of VEGF, the interaction with its receptors and its function in normal and pathological processes can be found in Hoeben et al. Pharmacol. Rev. 2004, 56: 549-580.
VEGF has been reported to be a pivotai regulator of both normal and abnormal angiogenesis (Ferrara and Davis-Smyth, Endocrine Rev. 1997,18:4-25; Ferrara J. MoL Med. 1999, 77: 527-543). Compared to other growth factors that contribute to the processes of vascular formation, VEGF is unique in its high specificity for endothélial cells within the vascular System.
VEGF mRNA is overexpressed by the majority of human tumors. In the case of tumor growth, angiogenesis appears to be crucial for the transition from hyperplasia to neoplasia, and for providing nourishment for the growth and metastasis of the tumor (Folkman étal., 1989, Nature 339-58), which allows the tumorcells to acquire a growth advantage compared to the normal cells. Therefore, anti-angiogenesis thérapies hâve become an important treatment option for several types of tumors. These thérapies hâve focused on blocking the VEGF pathway (Ferrara et al., Nat Rev Drug Discov. 2004 May; 3(5): 391-400.
VEGF is also involved in eye diseases. The concentration of VEGF in eye fluids is highly correlated with the presence of active prolifération of blood vessels in patients with diabetic and other ischemia-related rétinopathies. Furthermore, recent studies hâve demonstrated the localization of VEGF in choroidal neovascular membranes in patients affected by age-related macular degeneration (AMD). Up-regulation of VEGF has also been observed in various inflammatory disorders. VEGF has been implicated in the pathogenesis of rheutatoid arthritis, an inflammatory disease in which angiogenesis plays a significant rôle.
The élucidation of VEGF and its rôle in angiogenesis and different processes has provided a potential newtarget of therapeutic intervention. The function of VEGF has been inhibited by small molécules that block or prevent activation of VEGF receptor tyrosine kinases (Schlaeppi and Wood, 1999, Cancer Metastasis Rev., 18: 473-481) and consequently interfère with the VEGF receptor signal transduction pathway. Cytotoxic conjugates containing bacterial or plant toxins can inhibit the stimulating effect of VEGF on tumor angiogenesis. VEGF-DT385 toxin conjugates (diphtheria toxin domains fused or chemically conjugated to VEGF165), for example, efficiently inhibit tumor growth in vivo. Tumor growth inhibition could also be achieved by delivering a Flk-1 mutant or soluble VEGF receptors by a retrovirus.
VEGF-neutralizing antîbodies, such as A4.6.I and MV833, hâve been developed to block VEGF from binding to its receptors and hâve shown preclinical antitumor activity (Kim étal. Nature 1993, 362: 841-844; Folkman Nat. Med. 1995,1: 27-31; Presta et al. Cancer Res. 1997, 57: 4593-4599; Kanai et al. Int. J. Cancer 1998, 77: 933-936; Ferrara and Alitalo Nat. Med. 1999, 5: 1359-1364; 320, 340. For a review of therapeutic anti-VEGF approaches trials, see Campochiaro and Hackett (Oncogene 2003, 22: 6537-6548).
Most clinical expérience has been obtained with A4.6.1, also called bevacizumab (Avastin®; Genentech, San Francisco, CA).
W02008/101985 describes immunoglobulin single variable domains from camélidés (VHHs or Nanobodies®, as defined herein) that bind to VEGF, and their use in the treatment of conditions and diseases characterized by excessive and/or pathological angiogenesis or neovascularization.
It has been an object of the présent invention to provide novel anti-angiogenic io binding molécules for human therapy.
It has been a further object of the invention to provide methods for the prévention, treatment, aliénation and/or diagnosis of such diseases, disorders or conditions, involving the use and/or administration of such binding molécules and compositions comprising them. In particular, it is has been an object of the invention to provide is such pharmacologically active binding molécules, compositions and/or methods that provide advantages compared to the agents, compositions and/or methods currently used and/or known in the art. These advantages include improved therapeutic and/or pharmacological properties and/or other advantageous properties, e.g. for manufacturing purposes, especially as compared to conventional antibodies as those 20 described above, or fragments thereof.
BRIEF SUMMARY OF THE INVENTION
According to a first aspect, there are provided bispecific binding molécules, preferably bispecific immunoglobulins, preferably immunoglobulin single variable 25 domains like VHHs and domain antibodies, which comprise at least one VEGFbinding component and at least one Ang2-binding component in a single molécule. Preferably, said bispecific binding molécules further comprise a sérum albumin binding component.
More specifically, a bispecific binding molécule of the invention essentially comprises (i) a Ang2-binding component specifically binding to at least one epitope of Ang2 and (ii) a VEGF-binding component specifically binding to at least an epitope of VEGF, wherein the components are linked to each other in such a way that they simultaneously bind to Ang2 and VEGF or that they bind to either Ang2 or VEGF at a time.
According to preferred aspects of the invention, the two components comprise one or more immunoglobulin single variable domains that may be, independently of each 10 other, VHHs or domain antibodies, and/or any other sort of immunoglobulin single variable domains, such as VL domains, as defined herein, provided that each of these immunoglobulin single variable domains will bind the antigen, i.e. Ang2 or VEGF, respectively.
According to a preferred embodiment, the immunoglobulin single variable domains 15 are of the same type, in particular, ail immunoglobulin single variable domains are VHHs or domain antibodies.
According to a particularly preferred embodiment, ail immunoglobulin single variable domains are VHHs, preferably humanized (or sequence-optimized, as defined herein) VHHs. Accordingly, the invention relates to bispecific binding molécules comprising an (optionally humanized or sequence-optimized) anti-Ang2 VHH and an (optionally humanized or sequence-optimized) anti-VEGF VHH.
However, it will be clear to the skilled person that the teaching herein may be applied analogously to bispecific binding molécules including other anti-Ang2 or anti-VEGF immunoglobulin single variable domains, such as domain antibodies.
In another aspect, the invention relates to nucleic acids encoding the bispecific binding molécules of the invention as well as host cells containing same.
The invention further relates to a product or composition containing or comprising at least one bispecific binding molécule of the invention and optionally one or more further components of such compositions.
The invention further relates to methods for preparing or generating the bispecific binding molécules, nucleic acids, host cells, products and compositions described herein,
The invention further relates to applications and uses of the bispecific binding molécules, nucleic acids, host cells, products and compositions described herein, as well as to methods for the prévention and/or treatment for diseases and disorders that can be modulated by inhibition of Ang2.
It has been found that the Ang2-binding component of the bispecific binding molécules according to the présent invention binds to and antagonizes Ang2 with a potency at least 5,000 times higher, preferably 10,000 times higher than to Ang1 or Ang4. This will largely avoid blocking activation of Ang1 -mediated signalling, which would counter the intended anti-angiogenetic effect.
It has further been found that the VEGF-binding component of the bi-specific binding molécules according to the présent invention binds to VEGF-A with an affinity of at least 1,000 times higher, preferebly at least 5,000 times higher, more preferably at least 10,000 times higher than to VEGF-B, VEGF-C, VEGF-D or PLGF. Due to the highly preferential binding to VEGF-A the signaling of VEGFR3, which modulâtes of lymph angiogenesis, is not interfered with.
In a preferred embodiment the bispecific binding molécules of the présent invention are provided as linked VHH domains. Such molécules are significantly smallerthan conventional antibodies and hâve thus the potential for penetrating into a tumor deeper than such conventional antibodies. This benefit is further accentuated by the spécifie sequences disclosed herein after being free of glycosylation sites.
Further, due to the bispecific nature (VEGF- and Ang2-binding components in one molécule) the tumor pénétration of both functionalities will be necessarily equal, which will ensure that the bénéficiai effects of the combined antagonism of VEGF and Ang2 will be provided within the whole depth of pénétration of the tumor. This is an advantage over the combination of individual antagoniste against these targets, since the depth of pénétration of individual antagonists will always vary to some degree.
Another advantage of a preferred bispecific binding molécules of the présent invention is their increased sérum half-like due to a sérum albumin binding component such as a sérum albumin binding molécule as described herein.
These and other aspects, embodiments, advantages and applications of the invention will become clear from the further description hereinbelow.
DEFINITIONS
Unless indicated or defined otherwise, ail terms used hâve their usual meaning in the art, which will be clear to the skilled person. Reference is for example made to the standard handbooks, such as Sambrook et al, Molecular Cloning: A Laboratory Manual (2nd Ed.), Vols. 1-3, Cold Spring Harbor Laboratory Press (1989); Lewin, Genes IV, Oxford University Press, New York, (1990), and Roitt étal., Immunology (2nd Ed.), Gower Medical Publishing, London, New York (1989), as well as to the general background art cited herein; Furthermore, unless indicated otherwise, ail methods, steps, techniques and manipulations that are not specifically described in detail can be performed and hâve been performed in a manner known per se, as will be clear to the skilled person. Reference is for example again made to the standard handbooks, to the general background art referred to above and to the further references cited therein.
The term “bispecific binding moiecule” refers to a molécule comprising at least one Ang2-binding molecuie (or Ang2-binding component) and at least one VEGFbinding moiecule (or “VEGF-binding component). A bispecific binding molecuie may contain more than one Ang2-binding molecuie and/or more than one VEGF-binding moiecule, i.e. in the case that the bispecific binding moiecule contains a biparatopic (as defined below) Ang2-binding molécule and/or a biparatopic VEGF-binding molécule, in the part of the molécule that binds to Ang2 or to VEGF, i.e. in its Ang2binding component (or anti-Ang2 component) or “VEGF-binding component (or antiVEGF component), respectively. The word “bispecific” in this context is however not s to be construed as to exclude further binding components with binding specificity to molécules other than VEGF and Ang2 from the bispecific binding molécule. Nonlimiting examples of such further binding components are binding components binding to sérum albumin.
Unless indicated otherwise, the terms immunoglobulin and immunoglobulin sequence - whether used herein to refer to a heavy chain antibody or to a conventional 4-chain antibody - are used as general terms to include both the fullsize antibody, the individual chains thereof, as well as ail parts, domains or fragments thereof (including but not lîmited to antigen-binding domains or fragments such as VHH domains or VH/VL domains, respectively). In addition, the term sequence as is used herein (for example in terms like immunoglobulin sequence, antibody sequence, (single) variable domain sequence, VHH sequence or protein sequence), should generally be understood to include both the relevant amino acid sequence as well as nucleic acid sequences or nucléotide sequences encoding the same, unless the context requires a more lîmited interprétation.
The term domain (of a polypeptide or protein) as used herein refers to a folded protein structure which has the ability to retain its tertiary structure independently of the rest of the protein. Generally, domains are responsible for discrète functional properties of proteins, and in many cases may be added, removed or transferred to other proteins without loss of function of the remainder of the protein and/or of the domain.
The term immunoglobulin domain as used herein refers to a globular région of an antibody chain (such as e.g. a chain of a conventional 4-chain antibody or of a heavy chain antibody), or to a polypeptide that essentially consists of such a globular région. Immunoglobulin domains are characterized in that they retain the immunoglobulin fold characteristic of antibody molécules, which consiste of a 2-layer sandwich of about 7 antiparallel beta-strands arranged in two beta-sheets, optionally stabilized by a conserved disulphide bond. An immunoglobulin domain comprises (a) variable domain(s), i.e., one or more immunoglobulin variable domains.
The term ”immunoglobulin variable domain as used herein means an immunoglobulin domain essentially consisting of four framework régions which are referred to in the art and hereinbelow as framework région 1 or FR1; as framework région 2 orFR2; as framework région 3 or FR3; and as framework région 4 or FR4, respectively; which framework régions are interrupted by three complementarity determining régions or CDRs, which are referred to in the art and hereinbelow as complementarity determining région T'or CDR1; as complementarity determining région 2 or CDR2; and as complementarity determining région 3 or CDR3, respectively. Thus, the general structure or sequence of an immunoglobulin variable domain can be indicated as follows: FR1 CDR1 - FR2 - CDR2 - FR3 - CDR3 - FR4. It is the immunoglobulin variable domain(s) that confer specificity to an antibody for the antigen by carrying the antigen-binding site. In the context of the présent invention immunoglobulin single variable domains like VHHs and domain antibodies are preferred.
The term immunoglobulin single variable domain as used herein means an immunoglobulin variable domain which is capable of specifically binding to an epitope of the antigen without pairing with an additional variable immunoglobulin domain. One example of immunoglobulin single variable domains in the meaning of the présent invention are domain antibodies, such as the immunoglobulin single variable domains VH and VL (VH domains and VL domains). Another example of immunoglobulin single variable domains are VHH domains (or simply VHHs) from camelids, as defined hereinafter.
In view of the above définition, the antigen-binding domain of a conventional 4-chain antibody (such as an IgG, IgM, IgA, IgD or IgE molécule; known in the art) or of a Fab fragment, a F(ab')2 fragment, an Fv fragment such as a disulphide linked Fv or a scFv fragment, or a diabody (ail known in the art) derived from such conventîonal 4chain antibody, would normally not be regarded as an immunoglobulin single variable domain, as, in these cases, binding to the respective epitope of an antigen would normally not occur by one (single) immunoglobulin domain but by a pair of (associating) immunoglobulin domains such as light and heavy chain variable domains, i.e. by a VH-VL pair of immunoglobulin domains, which jointly bind to an epitope of the respective antigen.
VHH domains, also known as VHHs, VhH domains, VHH antibody fragments, and VHH antibodies, hâve originally been described as the antigen binding immunoglobulin (variable) domain of heavy chain antibodies (i.e. of antibodies devoid of light chains; Hamers-Casterman C, Atarhouch T, Muyldermans S, Robinson G, Hamers C, Songa EB, Bendahman N, Hamers R.: Naturally occurring antibodies devoid of light chains; Nature 363, 446-448 (1993)). The term VHH domain has been chosen in order to distinguish these variable domains from the heavy chain variable domains that are présent in conventîonal 4-chain antibodies (which are referred to herein as Vh domains or VH domains) and from the light chain variable domains that are présent in conventîonal 4-chain antibodies (which are referred to herein as V|_ domains or VL domains). VHH domains can specifically bind to an epitope without an additional antigen binding domain (as opposed to VH or VL domains in a conventîonal 4-chain antibody, in which case the epitope is recognized by a VL domain together with a VH domain). VHH domains are small, robust and efficient antigen récognition units formed by a single immunoglobulin domain.
In the context of the présent invention, the terms VHH domain, VHH, VhH domain, VHH antibody fragment, VHH antibody, as well as Nanobody® and Nanobody® domain (Nanobody being a trademark of the company Ablynx N.V.; Ghent; Belgium) are used interchangeably and are représentatives of immunoglobulin single variable domains (having the structure FR1-CDR1-FR2-CDR2-FR3-CDR3-FR4 and specifically binding to an epitope without requiring the presence of a second immunoglobulin variable domain), and which are distinguished from VH domains by the so-called hallmark residues, as defined in e.g. W02009/109635, Fig. 1.
The amino acid residues of a immunoglobulin single variable domain, e.g. a VHH, are numbered according to the general numbering for Vh domains given by Kabat et al.
(Sequence of proteins of immunological interest, US Public Health Services, NIH Bethesda, MD, Publication No. 91), as applied to VHH domains from Camelids, as shown e.g. in Figure 2 of Riechmann and Muyldermans, J. Immunol. Methods 231, 25-38 (1999). According to this numbering
- FR1 comprises the amino acid residues at positions 1-30, io - CDR1 comprises the amino acid residues at positions 31-35,
- FR2 comprises the amino acids at positions 36-49,
- CDR2 comprises the amino acid residues at positions 50-65,
- FR3 comprises the amino acid residues at positions 66-94,
- CDR3 comprises the amino acid residues at positions 95-102, and
- FR4 comprises the amino acid residues at positions 103-113.
However, it should be noted that - as is well known in the art for Vh domains and for VHH domains - the total number of amino acid residues in each of the CDRs may vary and may not correspond to the total number of amino acid residues indicated by the Kabat numbering (that is, one or more positions according to the Kabat numbering may not be occupied in the actual sequence, or the actual sequence may contain more amino acid residues than the number allowed for by the Kabat numbering). This means that, generally, the numbering according to Kabat may or may not correspond to the actual numbering of the amino acid residues in the actual sequence.
Alternative methods for numbering the amino acid residues of Vh domains, which methods can also be applied in an analogous manner to VHH domains, are known in the art. However, in the présent description, daims and figures, the numbering according to Kabat and applied to VHH domains as described above will be followed, unless indicated otherwise.
The total number of amino acid resîdues in a VHH domain will usually be in the range of from 110 to 120, often between 112 and 115. It should however be noted that smaller and longer sequences may also be suitable for the purposes described herein.
Immunoglobulin single variable domains, e.g. VHHs and domain antibodies, according to the preferred embodiments of the invention, hâve a number of unique structural characteristics and functional properties which makes them highly advantageous for use in therapy as functional antigen-binding molécules. In particular, and without being limited thereto, VHH domains (which hâve been w designed by nature to functionally bind to an antigen without pairing with a light chain variable domain) can function as single, relatively small, functional antigenbinding structural units.
Due to their unique properties, immunoglobulin single variable domains, as defined herein, like VHHs or VHs (or VLs) - either alone or as part of a larger polypeptide,
e.g, a biparatopic molécule - offer a number of significant advantages:
• only a single domain is required to bind an antigen with high affinity and with high selectivity, so that there is no need to hâve two separate domains présent, nor to assure that these two domains are présent in the right spacial conformation and configuration (i.e. through the use of especially designed linkers, as with scFv's);
• immunoglobulin single variable domains can be expressed from a single nucleic acid molécule and do not require any post-translational modification (like glycosylation;
• immunoglobulin single variable domains can easily be engineered into multivalent and multispecific formats (as further discussed herein);
• immunoglobulin single variable domains hâve high specificity and affinity for their target, low inhérent toxicity and can be administered via alternative routes than infusion or injection;
• immunoglobulin single variable domains are htghly stable to heat, pH, proteases and other denaturing agents or conditions and, thus, may be prepared, stored or transported without the use of réfrigération equipments;
• immunoglobulin single variable domains are easy and relatively inexpensive to préparé, both on small scale and on a manufacturing scale. For example, immunoglobulin single variable domains can be produced using microbial fermentation (e.g. as further described below) and do not require the use of mammalian expression Systems, as with for example conventional antibodies;
• immunoglobulin single variable domains are relatively small (approximately kDa, or 10 times smaller than a conventional IgG) compared to conventional 4-chain antibodies and antigen-binding fragments thereof, and therefore show high(er) pénétration into tissues (including but not limited to solid tumors and other dense tissues) and can be administered in higher doses than such conventional 4-chain antibodies and antigen-binding fragments thereof;
• VHHs hâve spécifie so-called “cavity-binding properties (inter alia due to their extended CDR3 loop, compared to VH domains from 4-chain antibodies) and can therefore also access targets and epitopes not accessible to conventional 4-chain antibodies and antigen-binding fragments thereof;
• VHHs hâve the particular advantage that they are highly soluble and very stable and do not hâve a tendency to aggregate (as with the mouse-derived antigen-binding domains described by Ward et al., Nature 341: 544-546 (1989)).
The immunoglobulin single variable domains of the invention are not limited with respect to a spécifie biological source from which they hâve been obtained or to a spécifie method of préparation. For example, obtaining VHHs may include the following steps:
(1 ) isolating the VHH domain of a naturally occurring heavy chain antibody; or screening a library comprising heavy chain antibodies or VHHs and isolating VHHs therefrom;
(2) expressing a nucleic acid molécule encoding a VHH with the naturally occurring sequence;
io (3) humanizing (as described herein) a VHH, optionally after affinity maturation, with a naturally occurring sequence or expressing a nucleic acid encoding such humanized VHH;
(4) camelizing (as described below) a immunoglobulin single variable heavy domain from a naturally occurring antibody from an animal species, in particular a species of mammal, such as from a human being, or expressing a nucleic acid molécule encoding such camelized domain;
(5) camelizing a VH, or expressing a nucleic acid molécule encoding such a camelized VH;
(6) using techniques for preparing synthetically or semi-synthetically proteins, polypeptides or other amino acid sequences;
(7) preparing a nucleic acid molécule encoding a VHH domain using techniques for nucleic acid synthesis, followed by expression of the nucleic acid thus obtained;
(8) subjecting heavy chain antibodies or VHHs to affinity maturation, to mutagenesis (e.g. random mutagenesis or site-directed mutagenesis) and/or any other technique(s) in order to increase the affinity and/or specificity of the VHH; and/or (9) combinations or sélections of the foregoing steps.
Suitable methods and techniques for performing the above-described steps are known in the art and will be clear to the skilled person. By way of example, methods of obtaining VHH domains binding to a spécifie antigen or epitope hâve been described in W02006/040153 and W02006/122786.
According to spécifie embodiments, the immunoglobulin single variable domains of the invention or présent in the polypeptides of the invention are VHH domains with an amino acid sequence that essentially corresponds to the amino acid sequence of a naturally occurring VHH domain, but that has been humanized or sequenceoptimized (optionally after affinity-maturation), i.e. by replacing one or more amino acid residues in the amino acid sequence of said naturally occurring VHH sequence by one or more of the amino acid residues that occur at the corresponding position(s) in a variable heavy domain of a conventional 4-chain antibody from a human being. This can be performed using methods known in the art, which can by routinely used by the skilled person.
A humanized VHH domain may contain one or more fully human framework région sequences, and, in an even more spécifie embodiment, may contain human framework région sequences derived from the human germline Vh3 sequences DP29, DP-47, DP-51, or parts thereof, or be highly homologous thereto, optionally combined with JH sequences, such as JH5. Thus, a humanization protocol may comprise the replacement of any of the VHH residues with the corresponding framework 1,2 and 3 (FRI, FR2 and FR3) residues of germline VH genes such as DP 47, DP 29 and DP 51) either alone or in combination. Suitable framework régions (FR) of the immunoglobulin single variable domains of the invention can be selected from those as set out e.g. in W02006/004678 and specifically, include the so-called KERE and GLEW classes. Examples are immunoglobulin single variable domains having the amino acid sequence G-L-E-W at about positions 44 to 47, and their respective humanized counterparts. A humanized VHH domain may contain one or more fully human framework région sequences.
By way of example, a humanizing substitution for VHHs belonging to the 103 P,R,Sgroup and/or the GLEW-group (as defined below) is 108Q to 108L. Methods for humanizing immunoglobulin single variable domains are known in the art.
Binding immunoglobulin single variable domains with improved properties in view of therapeutic application, e.g. enhanced affinity or decreased immunogenicity, may be obtained from individual binding molécules by techniques known in the art, such as affinity maturation (for example, starting from synthetic, random or naturally occurring immunoglobulin sequences), CDR grafting, humanizing, combining fragments derived from different immunoglobulin sequences, PCR assembly using overlapping primera, and similar techniques for engineering immunoglobulin sequences well known to the skilled person; or any suitable combination of any of the foregoing, also termed sequence optimization, as described herein. Reference is, for example, made to standard handbooks, as well as to the further description and Examples.
If appropriate, a binding molécule with increased affinity may be obtained by affînitymaturation of another binding molécule, the latter representing, with respect to the affinity-matured molécule, the “parent binding molécule.
Methods of obtaining VHHs that bind to a spécifie antigen or epitope hâve been described earlier, e.g. in W02006/040153 and W02006/122786. As also described therein in detail, VHH domains derived from camelids can be humanized (also termed sequence-optimized herein, sequence-optimizing” may, in addition to humanization, encompass an additional modification of the sequence by one or more mutations that furnish the VHH with improved properties, such as the removal of potential post translational modification sites) by replacing one or more amino acid residues in the amino acid sequence of the original VHH sequence by one or more of the amino acid residues that occur at the corresponding position(s) in a VH domain from a conventional 4-chain antibody from a human being. A humanized VHH domain can contain one or more fuliy human framework région sequences, and, in an even more spécifie embodiment, can contain human framework région sequences derived from DP-29, DP-47, DP-51, or parts thereof, optionally combined with JH sequences, such as JH5.
Domain antibodies, also known as Dabs and dAbs (the terms Domain Antibodies and dAbs being used as trademarks by the GlaxoSmithKIine group of s companies) hâve been described in e.g. Ward, E.S., et al.: Binding activities of a répertoire of single immunoglobulin variable domains secreted from Escherichia coli; Nature 341; 544-546 (1989); Holt, L.J. étal.: Domain antibodies: proteins for therapy; TRENDS in Biotechnology 21(11): 484-490 (2003); and W02003/002609.
Domain antibodies essentiaily correspond to the VH or VL domains of antibodies io from non-camelid mammals, in particular human 4-chain antibodies. In order to bind an epitope as a single antigen binding domain, i.e. without being paired with a VL or VH domain, respectively, spécifie sélection for such antigen binding properties is required, e.g. by using libraries of human single VH or VL domain sequences.
Domain antibodies hâve, like VHHs, a molecular weight of approximately 13 to is approximately 16 kDa and, if derived from fully human sequences, do not require humanization for e.g. therapeutical use in humans. As in the case of VHH domains, they are well expressed also in prokaryotic expression Systems, providing a significant réduction in overall manufacturing cost.
Furthermore, it will also be clear to the skilled person that it is possible to graft one or more of the CDR’s mentioned above onto other scaffolds, including but not limited to human scaffolds or non-immunoglobulin scaffolds. Suitable scaffolds and techniques for such CDR grafting are known in the art.
The terms epitope and antigenic déterminant', which can be used interchangeably, refer to the part of a macromolecule, such as a polypeptide, that is recognized by antigen-binding molécules, such as conventional antibodies or the polypeptides of the invention, and more particularly by the antigen-binding site of said molécules. Epitopes define the minimum binding site for an immunoglobulin, and thus represent the target of specificity of an immunoglobulin.
A polypeptide (such as an immunoglobulin, an antibody, an immunoglobulin single variable domain of the invention, or generally an antigen-binding molécule or a fragment thereof) that can bind to or specifically bind to”, that has affinity foi and/or that has specificity for a certain epitope, antigen or protein (or for at least one part, fragment or epitope thereof) is said to be againsf or directed againsf said epitope, antigen or protein or is a binding molécule with respect to such epitope, antigen or protein. In this context, a VEGF-binding component may also be referred to as “VEGF-neutralizing.
Generally, the term specificity refers to the number of different types of antigens or epitopes to which a particular antigen-binding molécule or antigen-binding protein (such as an immunoglobulin single variable domain of the invention) molécule can bind. The specificity of an antigen-binding molécule can be determined based on its affinity and/or avidity. The affinity, represented by the equilibrium constant for the dissociation of an antigen with an antigen-binding protein (KD), is a measure for the binding strength between an epitope and an antigen-binding site on the antigenbinding protein: the lesser the value of the KD, the stronger the binding strength between an epitope and the antigen-binding molécule (alternatively, the affinity can also be expressed as the affinity constant (KA), which is 1 /KD). As will be clear to the skilled person (for example on the basis of the further disclosure herein), affinity can be determined in a manner known per se, depending on the spécifie antigen of interest. Avidity is the measure of the strength of binding between an antigen-binding molécule (such as an immunoglobulin, an antibody, an immunoglobulin single variable domain or a polypeptides containing it and the pertinent antigen. Avidity is related to both the affinity between an epitope and its antigen binding site on the antigen-binding molécule and the number of pertinent binding sites présent on the antigen-binding molécule.
The part of an antigen-binding molécule that recognizes the epitope is called a paratope.
Unless indicated otherwise, the term VEGF-binding molécule” or Ang2-binding molécule” includes anti-VEGF or anti-Ang2 antibodies, anti-VEGF antibody or antiAng2 antibody fragments, anti-VEGF antibody-like molécules or “anti-Ang2 antibody-like molécules”, as defined herein, and conjugates with any of these.
Antibodies include, but are not limited to, monoclonal and chimerized monoclonal antibodies. The term „antibody“ encompasses complété immunoglobulins, like monoclonal antibodies produced by recombinant expression in host cells, as well as antibody fragments or antibody-like molécules”, including single-chain antibodies and linear antibodies, so-called SMIPs” (Small Modular Immunopharmaceuticals”), as e.g described in W02002/056910; Antibody-like molécules include immunoglobulin single variable domains, as defined herein. Other examples for antibody-like molécules are immunoglobulin super family antibodies (IgSF), or CDRgrafted molécules.
Ang2-binding molécule or “VEGF-binding molécule respectively, refers to both is monovalent target-binding molécules (i.e. molécules that bind to one epitope of the respective target) as well as to bi- or multivalent binding molécules (i.e. binding molécules that bind to more than one epitope, e.g. biparatopic” molécules as defined hereinbelow). Ang2(or VEGF)-binding molécules containing more than one Ang2(or VEGF)-binding immunoglobulin single variable domain are also termed “formatted” binding molécules, they may, within the target-binding component, in addition to the immunoglobulin single variable domains, comprise linkers and/or moieties with effector functions, e.g. half-life-extending moieties like albumin-binding immunoglobulin single variable domains, and/or a fusion partner like sérum albumin and/or an attached polymer like PEG.
The term biparatopic Ang2(or VE GF)-binding molécule or biparatopic immunoglobulin single variable domain as used herein shall mean a binding molécule comprising a first immunoglobulin single variable domain and a second immunoglobulin single variable domain as herein defined, wherein the two molécules bind to two non-overlapping epitopes of the respective antigen. The biparatopic binding molécules are composed of immunoglobulin single variable domains which hâve different specificities with respect to the epitope. The part of an antigen-binding molécule (such as an antibody or an immunoglobulin single variable domain of the invention) that recognizes the epitope is called a paratope.
A formatted binding molécule may, albeit less preferred, also comprise two identical immunoglobulin single variable domains or two different immunoglobulin single variable domains that recognize the same or overlapping epitopes or their respective antigen. In this case, with respect to VEGF, the two immunoglobulin single variable domains may bind to the same or an overlapping epitope in each of the two monomers that form the VEGF dimer.
Typically, the binding molécules of the invention will bind with a dissociation constant (Ko) of 10E-5 to 10E-14 moles/liter (M) or less, and preferably 10E-7 to 10E-14 moles/liter (M) or less, more preferably 10E-8 to 10E-14 moles/liter, and even more preferably 10E-11 to 10E-13, as measured e.g. in a Biacore or in a Kinexa assay), and/or with an association constant (Ka) of at least 10E7 ME-1, preferably at least
10E8 ME-1, more preferably at least 10E9 ME-1, such as at least 10E11 ME-1. Any Kd value greaterthan 10E-4 M is generally considered to indicate non-specific binding. Preferably, a polypeptide of the invention will bind to the desired antigen, i.e. VEGF or Ang2, respectively, with a KD less than 500 nM, preferably less than 200 nM, more preferably less than 10 nM, such as less than 500 pM. Spécifie binding of an antigen-binding protein to an antigen or epitope can be determined in any suitable manner known per se, including, for example, the assays described herein, Scatchard analysis and/or compétitive binding assays, such as radioimmunoassays (RIA), enzyme immunoassays (EIA) and sandwich compétition assays, and the different variants thereof known per se in the art.
Amino acid residues will be indicated according to the standard three-letter or oneletter amino acid code, as generally known and agreed upon in the art. When comparing two amino acid sequences, the term amino acid différence refers to insertions, délétions or substitutions of the indicated number of amino acid residues at a position of the référencé sequence, compared to a second sequence. In case of substitution(s), such substitution(s) will preferably be conservative amino acid substitution(s), which means that an amino acid residue is replaced with another amino acid residue of similar chemical structure and which has little or essentially no influence on the function, activity or other biological properties of the polypeptide. Such conservative amino acid substitutions are well known in the art, for example from W01998/49185, wherein conservative amino acid substitutions preferably are substitutions in which one amino acid within the following groups (i) - (v) is substituted by another amino acid residue within the same group: (i) small aliphatic, nonpolar or slightly polar residues: Ala, Ser, Thr, Pro and Gly; (ii) polar, negatively charged residues and their (uncharged) amides: Asp, Asn, Glu and Gin; (iii) polar, positively charged residues: His, Arg and Lys; (iv) large aliphatic, nonpolar residues: Met, Leu, Ile, Val and Cys; and (v) aromatic residues:
Phe, Tyr and Trp. Particularly preferred conservative amino acid substitutions are as follows: Ala into Gly or into Ser; Arg into Lys; Asn into Gin or into His; Asp into Glu;Cys into Ser; Gin into Asn; Glu into Asp; Gly into Ala or into Pro; His into Asn or into Gin; Ile into Leu or into Val; Leu into Ile or into Val; Lys into Arg, into Gin or into Glu; Met into Leu, into Tyr or into Ile; Phe into Met, into Leu or into Tyr; Ser into Thr; Thr into Ser;Trp into Tyr; Tyr into Trp or into Phe; Val into Ile or into Leu.
A polypeptide or nucleic acid molécule is considered to be (in) essentially isolated (form) ~ for example, when compared to its native biological source and/or the reaction medium or cultivation medium from which it has been obtained - when it has been separated from at least one other component with which it is usually associated in said source or medium, such as another protein/polypeptide, another nucleic acid, another biological component or macromolecule or at least one contaminant, impurity or minor component. In partîcular, a polypeptide or nucleic acid molécule is considered essentially isolated when it has been purified at least 2-fold, in partîcular at least 10-fold, more in partîcular at least 100-fold, and up to 1000-fold or more. A polypeptide or nucleic acid molécule that is in essentially isolated form Is preferably essentially homogeneous, as determined using a suitabie technique, such as a suitable chromatographical technique, such as polyacrylamide gel electrophoresis.
Sequence identitÿ' between two VEGF-binding molécule sequences or between two Ang2-binding molécule sequences indicates the percentage of amino acids that are identical between the sequences. It may be calculated or determined as described in paragraph f) on pages 49 and 50 of W02008/020079. Sequence similarity indicates the percentage of amino acids that either are identical or that represent conservative amino acid substitutions.
Alternative methods for numbering the amino acid residues of Vh domains, which methods can also be applied in an analogous mannerto VHH domains, are known in the art. However, in the présent description, daims and figures, the numbering according to Kabat and applied to VHH domains as described above will be followed, unless indicated otherwise.
An affinity-matured” VEGF-binding molécule or Ang2-binding molécule, in particular a VHH or a domain antibody, has one or more alterations in one or more CDRs which resuit in an improved affinity for VEGF or Ang2, as compared to the respective parent VEGF-binding molécule or Ang2-binding molécule. Afffinity-matured VEGF-binding molécules or Ang2-binding molécules of the invention may be prepared by methods known in the art, for example, as described by Marks étal., 1992, Biotechnology 10: 779-783, or Barbas, étal., 1994, Proc. Nat. Acad. Sci, USA 91: 3809-3813.; Shier et al., 1995, Gene 169:147-155; Yelton étal., 1995, Immunol. 155:1994-2004; Jackson étal., 1995, J. Immunol. 154(7):3310-9; and Hawkins étal., 1992, J. Mol. Biol. 226(3): 889 896; KS Johnson and RE Hawkins, Affinity maturation of antîbodies using phage display, Oxford University Press 1996.
For the présent invention, an “amino acid sequences of SEQ ID NO: x”: includes, if not otherwise stated, an amino acid sequence that is 100% identical with the sequence shown in the respective SEQ ID NO; x;
a) amino acid sequences that hâve at least 80% amino acid identity with the sequence shown in the respective SEQ ID NO: x;
b) amino acid sequences that hâve 3, 2, or 1 amino acid différences with the sequence shown in the respective SEQ ID NO: x.
The terms cancer and cancerous refer to or describe the physiological condition in mammals that is typically characterized by unregulated cell growth/proliferation. Exemples of cancer to be treated with a bispecific binding molécule of the invention, include but are not limited to carcinoma, lymphoma, blastoma, sarcoma, and leukemia. More particular examples of such cancers, as suggested for treatment with VEGF antagonists in US 2008/0014196, include squamous cell cancer, small-cell lung cancer, non-small cell lung cancer, adenocarcinoma of the lung, squamous carcinoma of the lung, cancer of the peritoneum, hepatocellular cancer, gastrointestinal cancer, pancreatic cancer, glioblastoma, cervical cancer, ovarian cancer, liver cancer, bladder cancer, hepatoma, breast cancer, colon cancer, colorectal cancer, endométrial or uterine carcinoma, salivary gland carcinoma, kidney cancer, liver cancer, prostate cancer, vulval cancer, thyroid cancer, hepatic carcinoma, gastric cancer, melanoma, and various types of head and neck cancer. Dysrégulation of angiogenesis can lead to many disorders that can be treated by compositions and methods of the invention. These disorders include both nonneoplastic and neoplastic conditions. Neoplasties include but are not limited those described above.
Non-neoplastic disorders include, but are not limited to, as suggested for treatment with VEGF antagonists in US2008/0014196, undesired or aberrant hypertrophy, arthritis, rheumatoid arthritis (RA), psoriasis, psoriatic plaques, sarcoidosis, atherosclerosis, atherosclerotic plaques, diabetic and other proliférative rétinopathies including retinopathy of prematurity, retrolental fibroplasia, neovascular glaucoma, age-related macular degeneration, diabetic macular edema, corneal neovascularization, corneal graft neovascularization, corneal graft rejection, retinal/choroidal neovascularization, neovascularization ofthe angle (rubeosis), ocular neovascular disease, vascular restenosis, arteriovenous malformations (AVM), meningioma, hemangioma, angiofibroma, thyroid hyperplasias (including Grave's disease), corneal and other tissue transplantation, chronic inflammation, lung inflammation, acute lung injury/ ARDS, sepsis, primary pulmonary hypertension, malignant pulmonary effusions, cérébral edema (e.g., associated with acute stroke/ closed head injury/ trauma), synovial inflammation, pannus formation in RA, myositis ossificans, hypertropic bone formation, osteoarthritis (OA), refractory ascites, polycystic ovarian disease, endometriosis, 3rd spacing of fluid diseases (pancreatitis, compartiment syndrome, burns, bowel disease), uterine fibroids, prématuré labor, chronic inflammation such as IBD (Crohn's disease and ulcerative colitis), rénal allograft rejection, inflammatory bowel disease, nephrotic syndrome, undesired or aberrant tissue mass growth (non-cancer), hémophilie joints, hypertrophie scars, inhibition of haïr growth, Osier-Weber syndrome, pyogénie granuloma retrolental fibroplasias, scleroderma, trachoma, vascular adhesions, synovitis, dermatitis, preeclampsia, ascites, pericardial effusion (such as that associated with pericarditis), and pleural effusion.
DETAILED DESCRIPTION OF THE INVENTION
In a first aspect, the présent invention relates to a bispecific binding molécule comprising at least one Ang2-binding component and at least one VEGF-binding component.
In a preferred embodiment, the présent invention relates to a bispecific binding molécule comprising at least one VEGF-binding component and at least one Ang2binding component which further comprises at least a further binding component, preferably a sérum albumin binding component (sérum albumin binding molécule).
In a preferred embodiment, the sérum albumin binding component of the binding molécule of the présent invention is an isolated immunoglobulin single variable domain or a polypeptide containing one or more of said immunoglobulin single variable domains, wherein said immunoglobulin single variable domain consists of four framework régions and three complementarity determining régions CDR1, CDR2 and CDR3, respectîvely, and wherein said CDR3 has an amino acid sequence selected from amino acid sequences shown in SEQ ID NOs: 257, 260, 263, 266, 269, 272, or 275.
More preferably, said one or more immunoglobulin single variable domain of the sérum albumin binding component contain
a. a CDR3 with an amino acid sequence selected from a first group of amino acid sequences shown in SEQ ID NOs: SEQ IDs NOs: 257, 260, 263, 266, 269, 272, or 275;
b. a CDR1 with an amino acid sequences selected from a second group of amino acid sequences shown SEQ ID NOs:255, 258, 261, 264, 267, 270, or 273;
c. a CDR2 with an amino acid sequences selected from a second group of amino acid sequences shown SEQ ID NOs:256, 259, 262, 265, 268, 271, or 274.
In a more preferred embodiment, said one or more immunoglobulin single variable domains of the sérum albumin binding component are VHHs, preferably having an amino acid sequence shown in SEQ ID NOs: 98 or 254.
According to preferred embodiments, said Ang2-binding component and said VEGFbinding component comprise at least one Ang2-binding immunoglobulin single variable domain and at least one VEGF-binding immunoglobulin single variable domain, respectîvely.
In a preferred aspect, said Ang2-binding component and said VEGF-binding component each comprise at least one VEGF-binding immunoglobulin single variable domain and at least one Ang2-binding immunoglobulin single variable domain, respectîvely, wherein each of said immunoglobulin single variable domains has four framework régions and three complementarity determining régions CDR1, CDR2 and CDR3, respectîvely.
Thus, the anti-Ang2 and/or the anti-VEGF component contained in the bispecific binding molécules of the invention may include two (or more) anti-Ang2 (or antiVEGF, respectively) immunoglobulin single variable domains, wherein the immunoglobulin single variable domains are directed against different epitopes within the Ang2 (or VEGF) target. Thus, the two immunoglobulin single variable domains in a bispecific binding molécule will hâve different antigen specificity and therefore different CDR sequences.
Such bivalent binding molécules are also named biparatopic single domain antibody constructs (if the immunoglobulin single variable domains consist or essentially consist of single domain antibodies), or biparatopic VHH constructs (if the immunoglobulin single variable domains consist or essentially consist of VHHs), respectively, as the two immunoglobulin single variable domains will include two different paratopes.
In the bispecific binding molécule of the invention, one or both of the binding molécules may be bivalent; e.g. the VEGF-binding component may be biparatopic and the Ang2-binding component may be one immunoglobulin single variable domain, or the VEGF-binding component may be one immunoglobulin single variable domain and the Ang2-binding component may be biparatopic.
In bispecific binding molécules of the invention, it is preferably the VEGF-binding component that contains a bivalent VEGF-binding immunoglobulin single variable domain, e.g. a biparatopic VHH.
Such VEGF-binding immunoglobulin single variable domain may be two or more VEGF-binding VHHs, which are
a. identical VHHs that are capable of blocking the interaction between recombinant human VEGF and the recombinant human VEGFR-2 with an inhibition rate of £ 60% or
b. different VHHs that bind to non-overlapping epitopes of VEGF, wherein at least one VHH is capable of blocking the interaction between recombinant human
VEGF and the recombinant human VEGFR-2 with an inhibition rate of > 60% and wherein at least one VHH is capable of blocking said interaction with an inhibition rate of S 60 %.
The VEGF-binding component comprising at least a variable domain with four framework régions and three complementarity determining régions CDR1, CDR2 and CDR3, respectively, wherein said CDR3 has the amino acid sequence Ser Arg Ala Tyr Xaa Ser Xaa Arg Leu Arg Leu Xaa Xaa Thr Tyr Xaa Tyr as shown in SEQ ID NO: 1, wherein
Xaa at position 5 is Gly or Ala;
Xaa at position 7 is Ser or Gly;
Xaa at position 12 is Gly, Ala or Pro;
Xaa at position 13 is Asp or Gly;
Xaa at position 16 is Asp or Glu; and wherein said VEGF-binding component is capable of blocking the interaction of human recombinant VEGF165 with the human recombinant VEGFR-2 with an inhibition rate of >60%.
According to preferred embodiments, Xaa at position 5 is Gly, Xaa at position 7 is Ser, Xaa at position 12 is Ala, and Xaa at position 13 is Asp.
In particular, said CDR3 has a sequence selected from
SEQ ID NO: 2 SRAYGSSRLRLGDTYDY,
SEQ ID NO: 3 SRAYGSSRLRLADTYDY;
SEQ ID NO: 4 SRAYGSSRLRLADTYEY;
SEQ ID NO: 5 SRAYGSGRLRLADTYDY;
SEQ ID NO: 6 SRAYASSRLRLADTYDY;
SEQ ID NO: 7 SRAYGSSRLRLPDTYDY;
SEQ ID NO: 8 SRAYGSSRLRLPGTYDY.
According to certain embodiments, a VEGF-binding component comprises one or more immunoglobulin single variable domains each containing
a. a CDR3 with an amino acid sequence selected from a first group of sequences shown in SEQ ID NO: 2 to 8;
b. a CDR1 and a CDR2 with an amino acid sequences that is contained, as indicated in Table 3, in a sequence selected from a second group of amino acid sequences shown in SEQ ID NOs: 9 to 46, wherein said second sequence contains the respective CDR3 selected according to a).
According to preferred embodiments, the immunoglobulin single variable domains are VHHs.
According to spécifie embodiments, the VHHs hâve amino acid sequences selected from sequences shown in SEQ ID NOs: 9 - 46.
According to another spécifie embodiment, the VHHs hâve amino acid sequences selected from SEQ ID NOs: 15, SEQ ID NO: 18 and SEQ ID NO: 25.
The invention also relates to VEGF-binding component that hâve been obtained by affinity maturation and/or sequence optimization of an above-defined VHH, e.g. to a VHH that has been obtained by sequence optimization of a VHH having an amino acid sequence shown in SEQ ID NO: 18. Examples are VHHs having amino acid sequences selected from sequences shown in SEQ ID NOs: 47 - 57.
According to certain embodiments, a VEGF-binding domain of the invention may be formatted, as herein defined, e.g. it may be biparatopic or comprise two identical immunoglobulin single variable domains. Such VEGF-binding components may comprise two or more VHHs, which are
a) identical VHHs that are capable of blocking the interaction between recombinant human VEGF and the recombinant human VEGFR-2 with an inhibition rate of s 60% or
b) different VHHs that bind to non-overlapping epitopes of VEGF, wherein at Ieast one VHH is capable of blocking the interaction between recombinant human VEGF and the recombinant human VEGFR-2 with an inhibition rate of s 60% and wherein at least one VHH binds is capable of blocking said interaction with an inhibition rate of £ 60 %.
The percentage of blocking said interaction at an inhibition rate of à 60% or £ 60 %, respectively, refers to an inhibition rate as determined by an Amplified Luminescent Proximity Homogeneous Assay (AlphaScreen®), a compétition ELISA, a plasmon résonance (SPR) based assay (Biacore®) as used in the Examples.
In the following, the ability of VHHs according to a) is also termed “receptor-blocking, while the ability of VHHs according to b) is also termed non-receptor-blocking”.
Preferably, the receptor-blocking VHHs hâve an inhibition rate of s 80%, more preferably 2:90%; the most preferred VHHs being complété receptor blockers, i.e. hâve an inhibition rate of 100 %.
A VEGF-binding component may contain two or more identical VHHs a) selected from VHHs having amino acid sequences shown in SEQ ID NOs: 9 - 46 or VHHs that hâve been obtained by affinity maturation and/or sequence optimization of such VHH. The VHH may be selected from VHHs having the amino acid shown in SEQ ID NO: 18 or SEQ ID NO: 47 - 57.
According to preferred embodiments, a formatted VEGF-binding component comprises two VHHs each having the amino acid sequence shown in SEQ ID NO: 57.
In formatted VEGF-binding components comprising two different VHHs
a) said one or more VHHs with an inhibition rate of s 60% are selected from
i. VHHs having an amino acid sequence selected from amino acid sequences shown in SEQ ID NOs: 9 - 46 or ii. VHHs that hâve been obtained by affinity maturation and/or sequence optimization of such VHHs, and wherein
b) said one or more VHHs with an inhibition rate of 5 60 % are selected from
i. SEQ ID NOs: 58-124 or ii. VHHs that hâve been obtained by affinity maturation and/or sequence optimization of such VH H.
According to preferred embodiments, two VHHs are contained in polypeptides with amino acid sequences shown in SEQ ID NOs: 128 - 168, separated by linker sequences as indicated in Table 15.
In a preferred VEGF-binding component VHH a) i. has an amino acid sequence shown in SEQ ID NO: 18 and VHH b) i. has an amino acid sequence shown in SEQ ID NO: 64.
In other preferred VEGF-binding components VHHs according to a) ii. are selected from VHHs having an amino acid sequence shown in SEQ ID NOs: 47 - 57 and VHHs according to b) ii. are selected from VHHs having an amino acid sequence shown in SEQ ID NOs: 125 -127 .
Particularly preferred is a biparatopic VEGF-binding component comprising two VHHs, one of them having the amino acid shown in SEQ ID NO: 57 and one of them having the amino acid shown in SEQ ID NO: 127.
The Ang2-binding component comprises at least a variable domain with four framework régions and three complementarity determining régions CDR1, CDR2 and CDR3, respectively, wherein said CDR3 has an amino acid sequence selected from amino acid sequences shown in SEQ ID NOs: 226, 229, 232, 235, 238, 241, 244, 247, 250, or 253.
In a second aspect, said Ang2-binding component is an isolated immunoglobuîin single variable domain or a polypeptide containing one or more of said immunoglobuîin single variable domains, wherein said immunoglobuîin single variable domain consists of four framework régions and three complementarity determining régions CDR1, CDR2 and CDR3, respectively, and wherein said CDR3 has an amino acid sequence selected from amino acid sequences shown in SEQ ID NOs: 226, 229, 232, 235, 238, 241, 244, 247, 250, or 253.
In a further aspect, said immunoglobulin single variable domain of the Ang2-binding component contains
a. a CDR3 with an amino acid sequence selected from a first group of amino acid sequences shown in SEQ ID NOs: SEQ IDs NOs: 226, 229, 232, 235, 238, 241,244, 247, 250, or 253 (see also Table 49);
b. a CDR1 with an amino acid sequences that is contained, as indicated in Table 36-A, 38-A, 41-A, or 45-A, as partial sequence in a sequence selected from a second group of amino acid sequences shown SEQ ID NOs: 224, 227, 230,
233, 236, 239, 242, 245, 248, or 251 (see also Table 49);
c. a CDR2 with an amino acid sequences that is contained, as indicated in Table 36-A, 38-A, 41-A, or45-A, as partial sequence in a sequence selected from a second group of amino acid sequences shown SEQ ID NOs: 225, 228, 231,
234, 237, 240, 243, 246, 249, or 252 (see also Table 49).
Preferably, the immunoglobulin single variable domain of the Ang2-binding component is a VHH, preferably having amino acid sequence selected from amino acid sequences shown in SEQ ID NOs: 214, 215, 216, 217, 218, 219, 220, 221, 222, or 223.
In another preferred embodiment, the immunoglobulin single variable domain of the Ang2-binding component has been obtained by affinity maturation or humanization of an immunoglobulin single variable domain as described herein.
Similarly, the présent invention also relates to a VHH which has been obtained by affinity maturation or humanization of a VHH of the Ang2-binding component as described herein.
The présent invention thus also relates to an Ang2-binding VH H with an amino acid sequence selected from acid sequences shown in SEQ ID NOs: 214, 215, 216, 217, 218, 219, 220, 221,222, or 223.
Suitable parent Ang2-binding components for affinity maturation are, by way of example, the above-described VHHs with amino acid sequences shown in SEQ ID NOs:214, 215, 216, 217, 21 i8, or219.
Accordingly, the invention also relates to Ang2-binding molécules that hâve been obtained by affinity maturation and/or sequence optimization of an above-defined VHH, e.g. to a VHH that has been obtained by sequence optimization of a VHH having an amino acid sequence shown as SEQ ID NOs: 217, 218, 219, 220, 221, 222, or 223. The source” amino acid sequences that were used to generate the latter VHHs are shown in SEQ ID NOs: 214, 215, or 216. Also these amino acid sequences are suitable Ang2-binding components that can be applied in the binding molécules of the présent invention.
As described herein, the binding molécule of the présent invention preferably comprises at least one sérum albumin binding component. Particularly preferred binding molécules thus hâve at least one VEGF-binding component, at least one Ang2-binding component and at least one sérum albumin binding component. The order of these three binding components could be any possible order such as the order set out in Table 36-B, 38-B, 40, 41-B, 42, 43, 45-B, 46-A, or 47-A; or in Figure 20, 23, 27, or 30, e.g. the VEGF-, Ang2-or sérum albumin binding component can be N-terminal or C-terminal. Notably, 1D01” (SEQ ID No: 214), Ί1Β07, “00027” (SEQ ID No:216), “00908, 7G08” (SEQ ID No:215), 00919”, “00921 (SEQ ID No: 220), 00928” (SEQ ID No:221), “00932, 00933”, 00934, 00935, “00936”, 00937, 00938” (SEQ ID No:222), or “00956” (SEQ ID No:223) as referred to in the legend of the aforementioned Tables and Figures stand for Ang2-binding components, while 00038” stands for a VEGF-binding component and “ALB11 stands for a sérum albumin binding component. None of them is to be construed to a spécifie sequence, but stands for a Ang2-, VEGF- and sérum albumin binding component in general when used in the context of possible set-ups of binding molécules of the présent invention.
However, it is preferred that the sérum albumin binding component is in between the 5 VEGF- and Ang2-binding component (or vice versa), while it is particularly preferred that at least one VEGF-binding component is N-terminal, followed by at least one sérum albumin binding component, followed by at least one Ang2-binding component at the C-Terminus. This set-up is shown to be specifically useful.
The présent invention relates thus in a preferred aspect to binding molécules 10 comprising at least one VEGF-binding component, at least one Ang2-binding component and at least one sérum albumin binding component having an amino acid sequence selected from the amino acid sequences shown in SEQ ID NOs: 180-213, “At least one binding component (VEGF, Ang2 or sérum albumin) when used herein includes that a binding molécule of the présent invention may contain one, two, three, is four or five VEGF-, Ang2-, and/or sérum albumin binding components (i.e., entities/units) which are preferably represented by an immunoglobulin singly variable domain as described herein.
The VEGF- and/or Ang2-binding components with improved properties in view of therapeutic application, e.g, enhanced affinity or decreased immunogenicity, may be 2o obtained from individual VEGF-or Ang2-binding components of the invention by techniques known in the art, such as affinity maturation (for example, starting from synthetic, random or naturally occurring immunoglobulin sequences), CDR grafting, humanizing, combining fragments derived from different immunoglobulin sequences, PCR assembly using overlapping primers, and similar techniques for engineering 25 immunoglobulin sequences well known to the skilled person; or any suitable combination of any of the foregoing, also termed sequence optimization”, as described herein. Référencé is, for example, made to standard handbooks, as well as to the further description and Examples.
If appropriate, a VEGF-or Ang2-binding component of the invention with increased affinity may be obtained by affinity-maturation of another VEGF-or Ang2-binding component, the latter representing, with respect to the affinity-matured molécule, the parent VEGF-binding component.
In VEGF or Ang2 VHHs of the invention that start with EVQ, the N-terminal E may be replaced by a D (which is often a resuit of sequence-optimization) or it may be missing (as for expression ofthe VHH in E.coli). Forformatted VEGF-binding components, this usually applies only to the VHH that is situated N-terminally.
A preferred, but non-limiting humanizing substitution for VEGF VHH domains belonging to the 103 P,R,S-group and/or the GLEW-group (as defined below) is 108Q to 108L. Methods for humanizing immunoglobulin single variable domains are known in the art.
According to another embodîment, the immunoglobulin single variable domain is a domain antibody, as defined herein.
In yet another embodîment, the représentatives of the class of VEGF-and/or Ang2binding immunoglobulin single variable domains ofthe invention hâve amino acid sequences that correspond to the amino acid sequence of a naturally occurring VH domain that has been camelized, i.e. by replacing one or more amino acid residues in the amino acid sequence of a naturally occurring variable heavy chain from a conventional 4-chain antibody by one or more amino acid residues that occur at the corresponding posîtion(s) in a VHH domain of a heavy chain antibody. This can be performed in a manner known per se, which will be ciear to the skilled person, and référencé is additionally be made to WO1994/04678. Such camelization may preferentially occur at amino acid positions which are présent at the VH-VL interface and at the so-called Camelidae Hallmark residues (see for example also WO1994/04678). A detailled description of such humanization and camelization techniques and preferred framework région sequences consistent therewith can additionally be taken from e.g. pp. 46 and pp. 98 of W02006/040153 and pp. 107 of W02006/122786.
The VEGF-binding components of the invention, e.g. immunoglobulin single variable domains, hâve specificity for VEGF in that they comprise one or more immunoglobulin single variable domains specifically binding to one or more epitopes within the VEGF molécule. The same is true for Ang2-binding components of the invention.
Spécifie binding of an VEGF-binding component to its antigen VEGF can be determined in any suitable manner known per se, including, for example, the assays described herein, Scatchard analysis and/or compétitive binding assays, such as radioimmunoassays (RIA), enzyme immunoassays (EIA and ELISA) and sandwich compétition assays, and the different variants thereof known per se in the art. The same is true for an Ang2-binding component when binding to its antigen.
With regard to the antigen VEGF, a VEGF-binding component of the invention, e.g. an immunoglobulin single variable domain, is not limited with regard to the species. Thus, the immunoglobulin single variable domains of the invention preferably bind to human VEGF, if intended for therapeutic purposes in humans. However, immunoglobulin single variable domains that bind to VEGF from another mammalian species are also within the scope of the invention. An immunoglobulin single variable domain of the invention binding to one species form of VEGF may cross-react with VEGF, which has a different sequence than the human one, from one or more other species. For example, immunoglobulin single variable domains of the invention binding to human VEGF may exhibit cross reactivity with VEGF from one or more other species of primates and/or with VEGF from one or more species of animais that are used in animal models for diseases, for example monkey, mouse, rat, rabbit, pig, dog, and in particular in animal models for diseases and disorders associated with VEGF-mediated effects on angiogenesis (such as the species and animal models mentioned herein). Immunoglobulin single variable domains of the invention that show such cross-reactivity are advantageous in a research and/or drug development, since it allows the immunoglobulin single variable domains of the invention to be tested in acknowledged disease models such as monkeys, in particular Cynomolgus or Rhésus, or mice and rats.
Preferably, in view of cross-reactlvity with one or more VEGF molécules from species other than human that is/are intended for use as an animal model during development of a therapeutic VEGF antagonist, a VEGF-binding component recognizes an epitope in a région of the VEGF of interest that has a high degree of identity with human VEGF.
An immunoglobulin single variable domain of the invention recognizes an epitope which is, totally or in part, located in a région of VEGF that is relevant for binding to its receptor, in particular to VEGFR-2, which has been shown to be the receptor whose activation is causally involved in the neovascularisation of tumors. According to preferred aspects, immunoglobulin single variable domains of the invention block VEGF receptor activation, in particular VEGFR-2 activation, at least partially, preferably substantially and most preferably totally.
As described above, the ability of a VEGF-binding component to block the interaction between VEGF and its receptors, in particular the VEGFR-2, can be determined by an Amplifîed Luminescent Proximity Homogeneous Assay (AlphaScreen®), a compétition ELISA, or a plasmon résonance (SPR) based assay (Biacore®), as described in the Examples.
Preferably, an immunoglobulin single variable domain of the invention binds to VEGF with an affinity less than 500 nM, preferably less than 200 nM, more preferably less than 10 nM, such as less than 500 pM (as determined by Surface Plasmon Résonance analysis, as described in Example 5.7). The same is true for an immunoglobulin single variable domain of the invention binds to angiopoietin.
Preferably, the immunoglobulin single variable domains of the invention hâve IC50 values, as measured in a compétition ELISA assay as described in Example 5.1. in the range of 10'6 to 10’1° moles/litre or less, more preferably in the range of 10'8 to 10’ 10 moles/litre or less and even more preferably in the range of 10‘9 to 10'1° moles/litre or less.
According to a non-limiting but preferred embodiment of the invention, VEGF-binding immunoglobulin single variable domains of the invention bind to VEGF with an dissociation constant (Ko) of 10*5 to 10'12 moles/liter (M) or less, and preferably 10'7 to 10'12 moles/liter (M) or less and more preferably 10‘8 to 10'12 moles/liter (M), and/or with an association constant (Ka) of at least 107 M’1, preferably at least 108 M'1, more preferably at least 109 M1, such as at least 1012 M’1; and in particular with a Kd less than 500 nM, preferably less than 200 nM, more preferably less than 10 nM, such as less than 500 pM. The Kd and Ka values of the immunoglobulin single variable domain of the invention against VEGF can be determined. The same is true for an Ang2-binding immunoglobulin single variable domain of the invention.
Biparatopic VEGF-binding components comprising two or more immunoglobulin single variable domains essentially consist of or comprise (i) a first immunoglobulin single variable domain specifically binding to a first epitope of VEGF and (ii) a second immunoglobulin single variable domain specifically binding to a second epitope of VEGF, wherein the first epitope of VEGF and the second epitope of VEGF are not identical epitopes. In other words, such polypeptide of the invention comprises or essentially consist of two or more immunoglobulin single variable domains that are directed against at least two non-overlapping epitopes présent in VEGF, wherein said immunoglobulin single variable domains are linked to each other in such a way that they are capable of simultaneously binding VEGF. In this sense, the polypeptide of the invention can also be regarded as a bivalent or multivalent immunoglobulin construct, and especially as a multivalent immunoglobulin single variable domain construct, in that the polypeptide contains at least two binding sites for VEGF. (Such constructs are also termed formatted VEGF binding molécules, e.g. formatted” VHHs). The same is true for biparatopic Ang2-binding components, mutatis mutandis.
Such VEGF-or Ang2-binding component of the invention includes (at least) two antiVEGF or Ang2 immunoglobulin single variable domains, respectively, wherein (the) two immunoglobulin single variable domains are preferably directed against nonoverlapping epitopes within the VEGF molécule or angiopoietin molécule, respectively. Thus, these two immunoglobulin single variable domains will hâve a different antigen specificity and therefore different CDR sequences. For this reason, such polypeptides of the invention will herein also be named biparatopic polypeptides, or biparatopic domain antibody constructs (if the immunoglobulin single variable domains consist or essentially consist of domain antibodies), or biparatopic VHH constructs (if the immunoglobulin single variable domains consist or essentially consist of VHHs), respectively, as the two immunoglobulin single variable domains will include two different paratopes.
If a polypeptide of the invention is a biparatopic molécule as defined herein, at least one of the immunoglobulin single variable domain components binds to an epitope is such that the interaction between recombinant human VEGF and recombinant humen VEGFR-2 is blocked at an inhibition rate of >80%. As has been shown in experiments of the invention, certain formatted molécules contain two VHHs that both block the VEGFR2 receptor at an inhibition rate of £80%. Certain VHHs of the invention block the VEGFR-2 at an inhibition rate of 100%, i.e. they are complété blockers.
In both cases, additional sequences and moieties may be présent within the VEGFbinding components of the invention, e.g. N-terminally, C-terminally, or located between the two immunoglobulin single variable domains, e.g. linker sequences and sequences providing for effector fonctions, as set out in more detail herein.
According to another, albeit less preferred embodiment, a VEGF-binding component of the invention may include more than two anti-VEGF immunoglobulin single variable domains, i.e. three, four or even more anti-VEGF VHHs. In this case, at least two of the anti-VEGF immunoglobulin single variable domains are directed against non-overlapping epitopes within the VEGF molécule, wherein any further immunoglobulin single variable domain may bind to any of the two non-overlapping epitopes and/or a further epitope présent in the VEGF molécule.
According to the invention, the two or more immunoglobulin single variable domains can be, independently of each other, VHHs or domain antibodies, and/or any other 5 sort of immunoglobulin single variable domains, such as VL domains, as defined herein, provided that these immunoglobulin single variable domains will bind the antigen, i.e. VEGF or angiopoietin, respectively.
The detailed description of the binding components is primarily provided for the VEGF-binding component. However, ail features and options outlined herein for the 10 VEGF-binding component also apply equivalently for the Ang2-binding component, mutatis mutandis.
According to preferred embodiments, the binding molécules présent in the bispecific binding molécules (the Ang2-binding molécules within the Ang2-binding component or the VEGF-binding molécules within the VEGF-binding component or the two 15 adjacent Ang2- and VEGF-binding components) may be connected with each other directly (i.e. without use of a linker) or via a linker. The linker is preferably a linker peptide and will be selected so as to allow binding of the two different binding molécules to each of non-overlapping epitopes of the targets, either within one and the same target molécule, or within two different molécules.
2o In the case of biparatopic binding molécules, sélection of linkers within the Ang2- or the VEGF-binding component will inter alia dépend on the epitopes and, specifically, the distance between the epitopes on the target to which the immunoglobulin single variable domains bind, and will be clear to the skilled person based on the disclosure herein, optionally after some limited degree of routine expérimentation.
Two binding molécules (two VHHs or domain antibodies or VHH and a domain antibody), or two binding components, may be linked to each other via an additional VHH or domain antibody, respectively (in such binding molécules, the two or more immunoglobulin single variable domains may be linked directly to said additional immunoglobulin single variable domain or via suitable linkers). Such an additional VHH or domain antibody may for example be a VHH or domain antibody that provides for an increased half-life. For example, the latter VHH or domain antibody may be one that is capable of binding to a (human) sérum protein such as (human) sérum albumin or (human) transferrin.
Alternatively, the two or more immunoglobulin single variable domains that bind to the respective target may be linked in sériés (either directly or via a suitable linker) and the additional VHH or domain antibody (which may provide for increased halflife) may be connected directly or via a linker to one of these two or more aforementioned immunoglobulin sequences.
Suitable linkers are described herein in connection with spécifie polypeptides of the invention and may - for example and without limitation - comprise an amino acid sequence, which amino acid sequence preferably has a length of 9 or more amino acids, more preferably at Ieast 17 amino acids, such as about 20 to 40 amino acids. However, the upper lïmit is not critical but is chosen for reasons of convenience regarding e.g. biopharmaceutical production of such polypeptides.
The linker sequence may be a naturally occurring sequence or a non-naturally occurring sequence. If used for therapeutic purposes, the linker is preferably nonimmunogenic in the subject to which the bispecific binding molécule of the invention is administered.
One useful group of linker sequences are linkers derived from the hinge région of heavy chain antibodies as described in WO1996/34103 and WO1994/04678.
Other examples are poly-alanine linker sequences such as Ala- Ala- Ala.
Further preferred examples of linker sequences are Gly/Ser linkers of different length such as (glyxsery)z linkers, including (gly4ser)3, (gly4ser)4, (gly4ser), (glysser), gly3, and (gly3ser2)3.
Some non-limiting examples of linkers are contained în bispecific binding molécules of the invention shown in Table 15 (SEQ ID NOs 128 - 168), e.g. the linkers
GGGGSGGGGSGGGGSGGGGSGGGGSGGGGSGGGGS (35GS; SEQ ID NO: 169);
s GGGGSGGGS (9GS; SEQ ID NO: 170);
GGGGSGGGGSGGGGSGGGGSGGGGSGGGGSGGGGSGGGGS (40GS; SEQ ID NO: 171).
If a formatted bispecific binding molecuie of the invention is modified by the attachment of a polymer, for example of a polyethylene glycol PEG (polyethylene w glycol) moiety, the linker sequence preferably includes an amino acid residue, such as a cysteine or a lysine, allowing such modification, e.g. PEGylation, in the linker région.
Examples of linkers useful for for PEGylation are:
GGGGCGGGS (“GS9,C5, SEQ ID NO: 172);
is GGGGCGGGGSGGGGSGGGGSGGGGS (“GS25.C5, SEQ ID NO:173)
GGGSGGGGSGGGGCGGGGSGGGGSGGG (GS27,C14, SEQ ID NO:174),
GGGGSGGGGSGGGGCGGGGSGGGGSGGGGSGGGGS (GS35,C15, SEQ ID NO:175), and
GGGGCGGGGSGGGGSGGGGSGGGGSGGGGSGGGGS (“GS35.C5”, SEQ ID 20 NO: 176).
Furthermore, the linker may also be a poly(ethylene glycol) moiety, as shown in e.g. W02004/081026.
In another embodiment, the immunoglobulin single variable domains are linked to each other via another moiety (optionally via one or two linkers), such as another polypeptide which, in a preferred but non-limiting embodiment, may be a further immunoglobulin single variable domain as described above. Such moiety may either be essentially inactive or may hâve a biological effect such as improving the desired properties of the polypeptide or may confer one or more additional desired properties to the polypeptide. For example, and without limitation, the moiety may improve the half-life of the protein or polypeptide, and/or may reduce its immunogenicity or improve any other desired property.
According to a preferred embodiment, a bispecific binding molécule of the invention includes, especially when intended for use or used as a therapeutic agent, a moiety which extends the half-life of the polypeptide of the invention in sérum or other body fluids of a patient. The term half-life is defined as the time it takes for the sérum concentration of the (modified) polypeptide to reduce by 50%, in vivo, for example due to dégradation of the polypeptide and/or clearance and/or séquestration by natural mechanisms.
More specifically, such half-life extending moiety can be covalently linked to or fused to an immunoglobulin single variable domain and may be, without limitation, an Fc portion, an albumin moiety, a fragment of an albumin moiety, an albumin binding moiety, such as an anti-albumin immunoglobulin single variable domain, a transferrin binding moiety, such as an anti-transferrin immunoglobulin single variable domain, a polyoxyalkylene molécule, such as a polyethylene glycol molécule, an albumin binding peptide or a hydroxyethyl starch (HES) dérivative.
In another embodiment, the bispecific binding molécule of the invention comprises a moiety which binds to an antigen found in blood, such as sérum albumin, sérum immunoglobulins, thyroxine-binding protein, fibrinogen or transferrin, thereby conferring an increased half-life in vivo to the resulting polypeptide ofthe invention. According to a specifically preferred embodiment, such moiety is an albumin-binding immunoglobulin and, especially preferred, an albumin-binding immunoglobulin single variable domain such as an albumin-binding VHH domain.
If intended for use in humans, such albumin-binding immunoglobulin single variable domain preferably binds to human sérum albumin and preferably is a humanized albumin-binding VHH domain.
Immunoglobulin single variable domains binding to human sérum albumin are known in the art and are described in further detail in e.g. W02006/122786. Specifically, useful albumin binding VHHs are ALB 1 and its humanized counterpart, ALB 8 (W02009/095489). Other albumin binding VHH domains mentioned in the above patent publication may, however, be used as well.
A specifically useful albumin binding VHH domain is ALB8 which consists of or contains the amino acid sequence shown in SEQ ID NO: 98 or 254.
According to a further embodiment of the invention, the two immunoglobulin single variable domains, in preferably VHHs, may be fused to a sérum albumin molécule, such as described e.g. in W02001/79271 and WO2003/59934. As e.g. described in W02001 /79271, the fusion protein may be obtained by conventîonal recombinant technology: a DNA molécule coding for sérum albumin, or a fragment thereof, is joined to the DNA coding for the bispecific binding molécule, the obtained construct is inserted into a plasmid suitable for expression in the selected host cell, e.g. a yeast cell like Pichia pastoris or a bacterial cell, and the host cell is then transfected with the fused nucléotide sequence and grown under suitable conditions. The sequence of a useful HSA is shown in SEQ ID NO: 99.
According to another embodiment, a half-life extending modification of a polypeptide of the invention (such modification also reducing immunogenicity of the polypeptide) comprises attachment of a suitable pharmacologically acceptable polymer, such as straight or branched chain poly(ethylene glycol) (PEG) or dérivatives thereof (such as methoxypoly(ethylene glycol) or mPEG). Generally, any suitable form of PEGylation can be used, such as the PEGylation used in the art for antibodies and antibody fragments (including but not limited to domain antibodies and scFv's); reference is made, for example, to: Chapman, Nat. Biotechnol., 54, 531-545 (2002); Veronese and Harris, Adv. Drug Deliv. Rev. 54, 453-456 (2003); Harris and Chess, Nat. Rev. Drug. Discov. 2 (2003); and W02004/060965.
Various reagents for PEGylation of polypeptides are also commercially available, for example from Nektar Therapeutics, USA, or NOF Corporation, Japan, such as the Sunbright® EA Sériés, SH Sériés, MA Sériés, CA Sériés, and ME Sériés, such as Sunbright® ME-100MA, Sunbright® ME-200MA, and Sunbright® ME-400MA.
Preferably, site-directed PEGylation is used, in particular via a cysteine-residue (see for example Yang étal., Protein Engineering 16, 761-770 (2003)). For example, for this purpose, PEG may be attached to a cysteine residue that naturally occurs in a polypeptide of the invention, a polypeptide of the invention may be modified so asto suitably introduce one or more cysteine residues for attachment of PEG, or an amino acid sequence comprising one or more cysteine residues for attachment of PEG may be fused to the N- and/or C-terminus of a polypeptide of the invention, ail using techniques of protein engineering known per se to the skilled person.
Preferably, for the polypeptides of the invention, a PEG is used with a molecular weight of more than 5 kDa, such as more than 10 kDa and less than 200 kDa, such as less than 100 kDa; for example in the range of 20 kDa to 80 kDa.
With regard to PEGylation, its should be noted that generally, the invention also encompasses any bispecific binding molécule that has been PEGylated at one or more amino acid positions, preferably in such a way that said PEGylation either (1 ) increases the half-life in vivo; (2) reduces immunogenicity; (3) provides one or more further bénéficiai properties known per se for PEGylation; (4) does not essentially affect the affinity of the polypeptide for its target (e.g. does not reduce said affinity by more than 50 %, and more preferably not by more than 10%, as determined by a suitable assay described in the art); and/or (4) does not affect any of the other desired properties of the bispecific binding molécules of the invention. Suitable PEGgroups and methods for attaching them, either specifically or non-specifically, will be clear to the skilled person. Various reagents for PEGylation of polypeptides are also commercially available, for example from Nektar Therapeutics, USA, or NOF
Corporation, Japan, such as the Sunbright® EA Sériés, SH Sériés, MA Sériés, CA Sériés, and ME Sériés, such as Sunbright® ME-100MA, Sunbright® ME-200MA, and Sunbright® ME-400MA.
According to an especially preferred embodiment of the invention, a PEGylated polypeptide of the invention includes one PEG moiety of linear PEG having a molecular weight of 40 kDa or 60 kDa, wherein the PEG moiety is attached to the polypeptide in a linker région and, specifially, at a Cys residue at position 5 of a GS9linker peptide as shown in SEQ ID NO:93, at position 14 of a GS27-linker peptide as shown in SEQ ID NO:95, or at position 15 of a GS35-linker peptide as shown in SEQ ID NO:96, or at position 5 of a 35GS-linker peptide as shown in SEQ ID NO:97.
A bispecific binding molécule of the invention may be PEGylated with one of the PEG reagents as mentioned above, such as Sunbright® ME-400MA, as shown in the following chemical formula:
o II
o.
CH3O-(CH2CH2O)n-CHaCHaCHaNHCtCHsJa-N
Bispecific binding molécules that contain linkers and/or half-life extending functional groups are shown in SEQ ID NO: 81 and in Figure 48.
According to another embodiment, the immunoglobulin single variable domains are domain antîbodies, as defined herein.
Immunoglobulin single variable domains présent in the bispecific binding molécules of the invention may also hâve sequences that correspond to the amino acid sequence of a naturally occurring VH domain that has been camelized, i.e. by replacing one or more amino acid residues in the amino acid sequence of a naturally occurring variable heavy chain from a conventional 4-chain antibody by one or more amino acid residues that occur at the corresponding position(s) in a VHH domain of a heavy chain antibody. This can be performed in a manner known per se, which will be clear to the skilled person, and reference is additionally be made to
WO1994/04678. Such camelization may preferentially occur at amino acid positions which are présent at the VH-VL interface and at the so-called Camelidae Hallmark residues (see for example also WO1994/04678). A detailled description of such humanization and camelization techniques and preferred framework région sequences consistent therewith can additionally be taken from e.g. pp. 46 and pp. 98 of W02006/040153 and pp. 107 ofW02006/122786.
The binding components hâve specificity for Ang2 or VEGF, respectively, in that they io comprise in a preferred embodiment one or more immunoglobulin single variable domains specifically binding to one or more epitopes within the Ang2 molécule or within the VEGF molécule, respectively.
Spécifie binding of a binding component to its antigen Ang2 or VEGF can be determined in any suitable manner known per se, including, for example, the assays is described herein, Scatchard analysis and/or compétitive binding assays, such as radioimmunoassays (RIA), enzyme immunoassays (EIA and ELISA) and sandwich compétition assays, and the different variants thereof known per se in the art.
With regard to the antigen Ang2 or VEGF, respectively, an immunoglobulin single variable domain is not limited with regard to the species. Thus, the immunoglobulin 20 single variable domains preferably bind to human Ang2 or to human VEGF, respectively, if intended for therapeutic purposes in humans. However, immunoglobulin single variable domains that bind to Ang2 or VEGF, respectively, from another mammalian species, or polypeptides containing them, are also within the scope of the invention. An immunoglobulin single variable domain binding to one 25 species form of Ang2 or VEGF may cross-react with the respective antigen from one or more other species. For example, immunoglobulin single variable domains binding to the human antigen may exhibit cross reactivity with the respective antigen from one or more other species of primates and/or with the antigen from one or more species of animais that are used in animal models for diseases, for example monkey (in particular Cynomolgus or Rhésus), mouse, rat, rabbit, pig, dog or) and in particular in animal models for diseases and disorders that can be modulated by inhibition of Ang2 (such as the species and animal models mentioned herein). Immunoglobulin single variable domains of the invention that show such crossreactivity are advantageous in a research and/or drug development, since it allows the immunoglobulin single variable domains of the invention to be tested in acknowledged disease models such as monkeys, in particular Cynomolgus or Rhésus, or mice and rats.
Also, the binding components are not limited to or defined by a spécifie domain or an antigenic déterminant ofthe antigen against which they are directed. Preferably, in view of cross-reactivity with one or more antigen molécules from species other than human that is/are intended for use as an animal model during development of a therapeutic Ang2/VEGF antagonist, a binding component recognizes an epitope in a région of the the respective antigen that has a high degree of identity with the human antigen. By way of example, in view of using a mouse model, an anti-Ang2 immunoglobulin single variable domain contained in the bispecific binding molécules of the invention recognizes an epitope which is, totally or in part, located within the FLD domain of Ang2, which shows a high identity between human and mouse.
Preferably, the VEGF-binding component binds to the VEGF isoforms VEGF165 and/or VEGF121.
In another aspect, the invention relates to nucleic acid molécules that encode bispecific binding molécules ofthe invention. Such nucleic acid molécules will also be referred to herein as nucleic acids of the invention and may also be in the form of a genetic construct, as defined herein. A nucleic acid of the invention may be genomic DNA, cDNA or synthetic DNA (such as DNA with a codon usage that has been specifically adapted for expression in the intended host cell or host organism). According to one embodiment ofthe invention, the nucleic acid ofthe invention is in essentially isolated form, as defined hereabove.
The nucleic acid of the invention may also be in the form of, may be présent in and/or may be part of a vector, such as for example a plasmid, cosmid or YAC. The vector may especially be an expression vector, i.e. a vector that can provide for expression of the bispecific binding molécule in vitro and/or in vivo (i.e. in a suitable host cell, host organism and/or expression System). Such expression vector generally comprises at least one nucleic acid of the invention that is operably linked to one or more suitable regulatory éléments, such as promoter(s), enhancer(s), terminator(s), and the like. Such éléments and their sélection in view of expression of a spécifie sequence in a spécifie host are common knowledge of the skilled person. Spécifie examples of regulatory éléments and other éléments useful or necessary for expressing bispecific binding molécules of the invention, such as promoters, enhancers, terminators, intégration factors, sélection markers, leader sequences, reporter genes, and the like, are disclosed e.g. on pp. 131 to 133 of W02006/040153.
The nucleic acids of the invention may be prepared or obtained in a manner known per se (e.g. by automated DNA synthesis and/or recombinant DNA technology), based on the information on the amino acid sequences for the polypeptides of the invention given herein, and/or can be isolated from a suitable natural source.
In another aspect, the invention relates to host cells that express or that are capable of expressing one or more bispecific binding molécules of the invention; and/or that contain a nucleic acid of the invention. According to a particularly preferred embodiment, said host cells are bacterial cells; other useful cells are yeast cells, fungal cells or mammalian cells.
Suitable bacterial cells include cells from gram-negative bacterial strains such as strains of Escherichia coli, Proteus, and Pseudomonas, and gram-positive bacterial strains such as strains of Bacillus, Streptomyces, Staphylococcus, and Lactococcus. Suitable fungal cell include cells from species of Trichoderma, Neurospora, and Aspergillus. Suitable yeast cells include cells from species of Saccharomyces (for example Saccharomyces cerevisiae), Schizosaccharomyces (for example
Schizosaccharomyces pombe), Pichia (for example Pichia pastoris and Pichia methanolica), and Hansenula.
Suitable mammalian cells include for example CHO cells, BHK cells, HeLa cells, COS cells, and the like. However, amphibian cells, insect cells, plant cells, and any other cells used in the art for the expression of heterologous proteins can be used as well.
The invention further provides methods of manufacturing a bispecific binding molécule of the invention, such methods generally comprising the steps of;
- culturing host cells comprising a nucleic acid capable of encoding a bispecific binding molécule under conditions that allow expression of the bispecific binding molécule of the invention; and
- recovering or isolating the polypeptide expressed by the host cells from the culture; and
- optionally further purifying and/or modifying and/or formulating the bispecific binding molécule of the invention.
For production on an industrial scale, preferred host organisms include strains of E. coli, Pichia pastoris, and S. cerevisiae that are suitable for large scale expression, production and fermentation, and in particular for large scale pharmaceutical expression, production and fermentation.
The choice of the spécifie expression system dépends in part on the requirement for certain post-translational modifications, more specifically glycosylation. The production of a bispecific binding molécule of the invention for which glycosylation is desired or required would necessitate the use of mammalian expression hosts that hâve the ability to glycosylate the expressed protein. In this respect, it will be clear to the skilled person that the glycosylation pattern obtained (i.e. the kind, number and position of residues attached) will dépend on the cell or cell line that is used for the expression.
Bispecific binding molécules of the invention may be produced eitherin a cell as set out above intracellullarly (e.g. in the cytosol, in the periplasma or in inclusion bodies) and then isolated from the host cells and optionally further purified; or they can be produced extracellularly (e.g. in the medium in which the host cells are cultured) and then isolated from the culture medium and optionally further purified.
Methods and reagents used for the recombinant production of polypeptides, such as spécifie suitable expression vectors, transformation or transfection methods, sélection markers, methods of induction of protein expression, culture conditions, and the like, are known in the art. Similarly, protein isolation and purification techniques useful in a method of manufacture of a polypeptide of the invention are well known to the skilled person.
In a further aspect, the invention relates to a peptide having an amino acid sequence of a CDR3 contained in an anti-VEGF-VHH having an amino acid sequence selected from sequences shown in SEQ ID NOs: 9 to 57 or SEQ ID NOs: 58 -127, respectively, and a nucleic acid molécule encoding same.
These peptides correspond to CDR3s derived from the VHHs of the invention. They, in particular the nucleic acid molécules encoding them, are useful for CDR grafting in order to replace a CDR3 in an immunoglobulin chain, or for insertion into a nonimmunoglobulin scaffold, e.g. a protease inhibitor, DNA-binding protein, cytochrome b562, a helix-bundle protein, a disulfide-bridged peptide, a lipocalin or an anticalin, thus conferring target-binding properties to such scaffold. The method of CDRgrafting is well known in the art and has been widely used, e.g. for humanizing antibodies (which usually comprises grafting the CDRs from a rodent antibody onto the Fv frameworks of a human antibody).
In order to obtain an immunoglobulin or a non-immunoglobulin scaffold containing a CDR3 of the invention, the DNA encoding such molécule may be obtalned according to standard methods of molecular biology, e.g. by gene synthesis, by oligonucleotide annealing or by means of overlapping PCR fragments, as e.g. described by Daugherty étal., 1991, Nucleic Acids Research, Vol. 19, 9, 2471 -2476. A method for inserting a VHH CDR3 into a non-immunoglobulin scaffold has been described by Nicaise et al., 2004, Protein Science, 13,1882 - 1891.
The invention further relates to a product or composition containing or comprising at least one bispecific binding molécule of the invention and optionally one or more further components of such compositions known per se, i.e. depending on the intended use of the composition.
For pharmaceutical use, a bispecific binding molécule of the invention may be formulated as a pharmaceutical préparation or composition comprising at least one bispecific binding molécule of the invention and at least one pharmaceutically acceptable carrier, diluent or excipient and/or adjuvant, and optionally one or more further pharmaceutically active polypeptides and/or compounds. By means of nonlimiting examples, such a formulation may be in a form suitable for oral administration, for parentéral administration (such as by intravenous, intramuscular or subcutaneous injection or intravenous infusion), for topical administration, for administration by inhalation, by a skin patch, by an implant, by a suppository, etc. Such suitable administration forms - which may be solid, semi-solid or liquid, depending on the manner of administration - as well as methods and carriers for use in the préparation thereof, will be clear to the skilled person, and are further described herein.
Thus, in a further aspect, the invention relates to a pharmaceutical composition that contains at least one bispecific binding molécule, in particular one immunoglobulin single variable domain, of the invention and at least one suitable carrier, diluent or excipient (i.e. suitable for pharmaceutical use), and optionally one or more further active substances.
The bispecific binding molécules of the invention may be formulated and administered in any suitable manner known per se: Reference, in particular for the immunoglobulin single variable domains, is for example made to W02004/041862, W02004/041863, W02004/041865, W02004/041867 and W02008/020079, as well as to the standard handbooks, such as Remington’s Pharmaceutical Sciences, 18th
Ed., Mack Publishing Company, USA (1990), Remington, the Science and Practice of Pharmacy, 21lh Edition, Lippincott Williams and Wilkins (2005); or the Handbook of Therapeutic Antibodies (S. Dubel, Ed.), Wiley, Weinheim, 2007 (see for example pages 252-255).
For example, an immunoglobulin single variable domain of the invention may be formulated and administered in any manner known per se for conventional antibodies and antibody fragments (including ScFv’s and diabodies) and other pharmaceutically active proteins. Such formulations and methods for preparing the same will be clear to the skilled person, and for example include préparations suitable for parentéral administration (for example intravenous, intraperitoneal, subcutaneous, intramuscular, intraluminal, intra-arterial or intrathecal administration) or for topical (i.e. transdermal or intradermal) administration.
Préparations for parentéral administration may for example be stérile solutions, suspensions, dispersions or émulsions that are suitable for infusion or injection.
is Suitable carriers or diluents for such préparations for example include, without limitation, stérile water and pharmaceutically acceptable aqueous buffers and solutions such as physiological phosphate-buffered saline, Ringer’s solutions, dextrose solution, and Hank's solution; water oils; glycerol; éthanol; glycols such as propylene glycol or as well as minerai oils, animal oils and vegetable oils, for example peanut oil, soybean oil, as well as suitable mixtures thereof. Usually, aqueous solutions or suspensions will be preferred.
Thus, the bispecific binding molécule of the invention may be systemically administered, e.g., orally, in combination with a pharmaceutically acceptable vehicle such as an inert diluent or an assimilable edible carrier. For oral therapeutic administration, the bispecific binding molécule of the invention may be combined with one or more excipients and used in the form of ingestible tablets, buccal tablets, troches, capsules, élixirs, suspensions, syrups, wafers, and the like. Such compositions and préparations should contain at least 0.1% of the binding molécule of the invention. Their percentage in the compositions and préparations may, of course, be varied and may conveniently be between about 2 to about 60% of the weight of a given unit dosage form. The amount of the bispecific binding molécule of the invention in such therapeutically useful compositions is such that an effective dosage level will be obtained.
The tablets, pills, capsules, and the like may also contain binders, excipients, disintegrating agents, lubricants and sweetening or flavouring agents, for example those mentioned on pages 143-144 of W02008/020079. When the unit dosage form is a capsule, it may contain, in addition to materials of the above type, a liquid carrier, such as a vegetable oil or a polyethylene glycol. Various other materials may be présent as coatings or to otherwise modify the physical form of the solid unit dosage form. For instance, tablets, pills, or capsules may be coated with gelatin, wax, shellac or sugar and the like. A syrup or élixir may contain the binding molécules of the invention, sucrose or fructose as a sweetening agent, methyl and propylparabens as preservatives, a dye and flavoring such as cherry or orange flavor. Of course, any material used in preparing any unit dosage form should be pharmaceutically acceptable and substantially non-toxic in the amounts employed. In addition, the bispecific binding molécules of the invention may be incorporated into sustainedrelease préparations and devices.
Préparations and formulations for oral administration may also be provided with an enteric coating that will allow the constructs of the invention to resist the gastric environment and pass into the intestines. More generally, préparations and formulations for oral administration may be suitably formulated for delivery into any desired part of the gastrointestinal tract. In addition, suitable suppositories may be used for delivery into the gastrointestinal tract.
The bispecific binding molécules of the invention may also be administered intravenously or intraperitoneally by infusion or injection, as further described on pages 144 and 145 of W02008/020079.
For topical administration of the bispecific binding molécules of the invention, it will generally be désirable to administer them to the skin as compositions or formulations, in combination with a dermatologically acceptable carrier, which may be a solid or a liquid, as further described on page 145 of W02008/020079.
Generally, the concentration of the bispecific binding molécules of the invention in a liquid composition, such as a lotion, will be from about 0.1-25 wt-%, preferably from about 0.5-10 wt-%. The concentration in a semi-solid or solid composition such as a gel or a powder will be about 0.1-5 wt-%, preferably about 0.5-2.5 wt-%.
The amount of the bispecific binding molécules of the invention required for use in treatment will vary not only with the particular binding molécule selected, but also with the route of administration, the nature of the condition being treated and the âge and condition of the patient and will be ultimately at the discrétion of the attendant physician or clinician. Also, the dosage of the binding molécules of the invention varies depending on the target cell, tumor, tissue, graft, or organ.
The desired dose may conveniently be presented in a single dose or as divided doses administered at appropriate intervals, for example, as two, three, four or more sub-doses per day. The sub-dose itself may be further divided, e.g., into a number of discrète loosely spaced administrations; such as multiple inhalations from an insufflator or by application of a plurality of drops into the eye.
An administration regimen may include long-term, daily treatment. By “long-term is meant at least two weeks and preferably, several weeks, months, or years of duration. Necessary modifications in this dosage range may be determined by one of ordinary skill in the art using only routine expérimentation given the teachings herein. See Remington's Pharmaceutical Sciences (Martin, E.W., ed. 4), Mack Publishing Co., Easton, PA. The dosage can also be adjusted by the individual physician in the event of any complication.
According to a further embodiment, the invention relates to the use of bispecific binding molécules, e.g. immunoglobulin single variable domains, for therapeutic purposes, such as
- for the prévention, treatment and/or alleviation of a disorder, disease or condition, especially in a human being, that is associated with VEGF- and / or Ang2mediated effects on angiogenesis or that can be prevented, treated or alleviated by modulating the Notch signaling pathway and/or the Tie2 signalling pathway with a bispecific binding molécule according to the invention,
- in a method of treatment of a patient in need of such therapy, such method comprising administering, to a subject in need thereof, a pharmaceutically active amount of at least one bispecific binding molécule of the invention, e.g. an immunoglobulin single variable domain, or a pharmaceutical composition containing same;
- for the préparation of a médicament for the prévention, treatment or alleviation of disorders, diseases or conditions associated with VEGF- and/or Ang2-mediated effects on angiogenesis;
- as an active ingrédient in a pharmaceutical composition or médicament used for the above purposes.
According to a spécifie aspect, said disorder disorder, disease or condition is a cancer or cancerous disease, as defined herein.
According to another aspect, the disease is an eye disease associated with VEGFand/or Ang2-mediated effects on angiogenesis or which can be treated or alleviated by modulating the Notch signaling pathway with a bispecific binding molécule.
Depending on the cancerous disease to be treated, a bispecific binding molécule of the invention may be used on its own or in combination with one or more additional therapeutic agents, in particular selected from chemotherapeutic agents like DNA damaging agents or therapeutically active compounds that inhibit angiogenesis, signal transduction pathways or mitotic checkpoints in cancer cells.
The additional therapeutic agent may be administered simultaneously with, optionally as a component of the same pharmaceutical préparation, or before or after administration ofthe binding molécule.
In certain embodiments, the additional therapeutic agent may be, without limitation s (and in the case of the receptors, including the respective ligands), one or more inhibitors selected from the group of inhibitors of EGFR, VEGFR, HER2-neu, Her3, AuroraA, AuroraB, PLK and PI3 kinase, FGFR, PDGFR, Raf, Ras, KSP, PDK1, PTK2, IGF-R or IR.
Further examples of additional therapeutic agents are inhibitors of CDK, Akt, src/bcr 10 abl, cKit, cMet/HGF, c-Myc, Flt3, HSP90, hedgehog antagonists, inhibitors of
JAK/STAT, MEK, mTor, NFkappaB, the protéasome, Rho, an inhibîtor of wnt signaling or an inhibîtor of the ubiquitination pathway or another inhibîtor of the Notch signaling pathway.
Examples for Aurora inhibitors are, without limitation, PHA-739358, AZD-1152,
AT 9283, CYC-116, R-763, VX-680, VX-667, MLN-8045, PF-3814735.
An example for a PLK inhibîtor is GSK-461364.
Examples for raf inhibitors are BAY-73-4506 (also a VEGFR inhibîtor), PLX 4032, RAF-265 (also in addition a VEGFR inhibîtor), sorafenib (also in addition a VEGFR inhibîtor), and XL 281.
Examples for KSP inhibitors are ispinesib, ARRY-520, AZD-4877, CK-1122697, GSK246053A, GSK-923295, MK-0731, and SB-743921.
Examples for a src and/or bcr-abl inhibitors are dasatinib, AZD-0530, bosutinib, XL 228 (also an IGF-1R inhibîtor), nilotinib (also a PDGFR and cKit inhibîtor), imatinib (also a cKit inhibîtor), and NS-187.
An example for a PDK1 inhibîtor is BX-517.
An example for a Rho inhibîtor is BA-210.
Examples for PI3 kinase inhibitors are PX-866, BEZ-235 (also an mTor inhibitor), XL 418 (also an Akt inhibitor), XL-147, and XL 765 (also an mTor inhibitor).
Examples for inhibitors of cMet or HGF are XL-184 (also an inhibitor of VEGFR, cKit, Flt3), PF-2341066, MK-2461, XL-880 (also an inhibitor of VEGFR), MGCD-265 (also an inhibitor of VEGFR, Ron, Tie2), SU-11274, PHA-665752, AMG-102, and AV-299.
An example for a c-Myc inhibitor is CX-3543.
Examples for Flt3 inhibitors are AC-220 (also an inhibitor of cKit and PDGFR),
KW 2449, iestau rti ni b (also an inhibitor of VEGFR, PDGFR, PKC), TG-101348 (also an inhibitor of JAK2), XL-999 (also an inhibitor of cKit, FGFR, PDGFR and VEGFR), sunitinib (also an inhibitor of PDGFR, VEGFR and cKit), and tandutinib (also an inhibitor of PDGFR, and cKit).
Examples for HSP90 inhibitors are tanespimycin, alvespimycin, IPI-504 and CNF 2024.
Examples for JAK/STAT inhibitors are CYT-997 (also interacting with tubulin), TG 101348 (also an inhibitor of Flt3), and XL-019.
Examples for MEK inhibitors are ARRY-142886, PD-325901, AZD-8330, and XL 518.
Examples for mTor inhibitors are temsirolimus, AP-23573 (which also acts as a VEGF inhibitor), everolimus (a VEGF inhibitor in addition). XL-765 (also a PI3 kinase inhibitor), and BEZ-235 (also a PI3 kinase inhibitor).
Examples for Akt inhibitors are perifosine, GSK-690693, RX-0201, and triciribine.
Examples for cKit inhibitors are AB-1010, OSI-930 (also acts as a VEGFR inhibitor), AC-220 (also an inhibitor of Flt3 and PDGFR), tandutinib (also an inhibitor of Flt3 and PDGFR), axitinib (also an inhibitor of VEGFR and PDGFR), XL-999 (also an inhibitor of Flt3, PDGFR, VEGFR, FGFR), sunitinib (also an inhibitor of Flt3, PDGFR, VEGFR), and XL-820 (also acts as a VEGFR- and PDGFR inhibitor), imatinib (also a bcr-abl inhibitor), nilotinib (also an inhibitor of bcr-abl and PDGFR).
Examples for hedgehog antagonists are IPI-609 and CUR-61414.
Examples for CDK Inhibitors are seliciclib, AT-7519, P-276, ZK-CDK (also inhibiting VEGFR2 and PDGFR), PD-332991, R-547, SNS-032, PHA-690509, and AG 024322.
Examples for protéasome inhibitors are bortezomib, carfilzomib, and NPI-0052 (also an inhibitor of NFkappaB).
An example for an NFkappaB pathway inhibitor is NPI-0052.
An example for an ubiquitination pathway inhibitor is HBX-41108.
In preferred embodiments, the additional therapeutic agent is an anti-angiogenic agent.
Examples for anti-angiogenic agents are inhibitors of the FGFR, PDGFR and VEGFR or the respective ligands (e.g VEGF inhibitors like pegaptanib or the anti-VEGF antibody bevacizumab), EGFL7 inhibitors, such as anti-EGFL7 MAb , angiopoietin1/2 inhibitors such as AMG386, and thalidomides, such agents being selected from, without limitation, bevacizumab, motesanib, CDP-791, SU-14813, telatinib, KRN-951, ZK-CDK (also an inhibitor of CDK), ABT-869, BMS-690514, RAF-265, IMC-KDR, IMC-18F1, IMiDs (immunomodulatory drugs), thalidomide dérivative CC-4047, lenalidomide, ENMD 0995, IMC-D11, Ki 23057, brivanib, cediranib, XL-999 (also an inhibitor of cKit and Flt3), 1B3, CP 868596, IMC 3G3, R-1530 (also an inhibitor of Flt3), sunitinib (also an inhibitor of cKit and Flt3), axitinib (also an inhibitor of cKit), lestaurtinib (also an inhibitor of Flt3 and PKC), vatalanib, tandutinib (also an inhibitor of Flt3 and cKit), pazopanib, GW 786034, PF-337210, IMC-1121B, AVE-0005, AG13736, E-7080, CHIR 258, sorafenib tosylate (also an inhibitor of Raf), RAF-265 (also an inhibitor of Raf), vandetanib, CP-547632, OSI-930, AEE-788 (also an inhibitor of EGFR and Her2), BAY-57-9352 (also an inhibitor of Raf), BAY-73-4506 (also an inhibitor of Raf), XL 880 (also an inhibitor of cMet), XL-647 (also an inhibitor of EGFR and EphB4), XL 820 (also an inhibitor of cKit), and nilotinib (also an inhibitor of cKit and brc-abl).
The additional therapeutic agent may also be selected from EGFR inhibitors, it may be a small molécule EGFR inhibitor or an anti-EGFR antibody. Examples for antiEGFR antibodies, without limitation, are cetuximab, panitumumab, matuzumab; an example for a small molécule EGFR inhibitor is gefitinib. Another example for an 5 EGFR modulator is the EGF fusion toxin.
Among the EGFR and Her2 inhibitors useful for combination with the bispecific binding molécule of the invention are lapatinib, gefitinib, erlotinib, cetuximab, trastuzumab, nimotuzumab, zalutumumab, vandetanib (also an inhibitor of VEGFR), pertuzumab, XL-647, HKI-272, BMS-599626 ARRY-334543, AV 412, mAB-806,
BMS-690514, JN J-26483327, AEE-788 (also an inhibitor of VEGFR), ARRY-333786,
IMC-11F8, Zemab.
Other agents that may be advantageously combined in a therapy with the bispecific binding molécule of the invention are tositumumab and ibritumomab tiuxetan (two radiolabelled anti-CD20 antibodies), alemtuzumab (an anti-CD52 antibody), denosumab, (an osteoclast différentiation factor ligand inhibitor), galiximab (a CD80 antagonist), ofatumumab (a CD20 inhibitor), zanolimumab (a CD4 antagonist), SGN40 (a CD40 ligand receptor modulator), rituximab (a CD20 inhibitor), mapatumumab (a TRAIL-1 receptor agonist), REGN421(SAR153192) or OMP21M18 (DII4 inhibitors).
Other chemotherapeutic drugs that may be used in combination with the bispecific binding molécule of the présent invention are selected from, but not limited to hormones, hormonal analogues and antihormonals (e.g. tamoxifen, toremifene, raloxifene, fulvestrant, megestrol acetate, flutamide, nilutamide, bicalutamide, cyproterone acetate, finasteride, buserelin acetate, fludrocortisone, fluoxymesterone, medroxyprogesterone, octreotide, arzoxifene, pasireotide, vapreotide), aromatase inhibitors (e.g. anastrozole, letrozole, liarozole, exemestane, atamestane, formestane), LHRH agonists and antagonists (e.g. goserelin acetate, leuprolide, abarelix, cetrorelix, deslorelin, histrelin, triptorelin), antimetabolites (e.g. antifolates like methotrexate, pemetrexed, pyrimidine analogues like 5 fluorouracil, capecitabine, decitabîne, nelarabine, and gemcitabine, purine and adenosine analogues such as mercaptopurine thioguanine, cladribine and pentostatin, cytarabine, fludarabine); antitumor antibiotics (e.g. anthracyclines like doxorubicin, daunorubicin, epirubicin and idarubicin, mitomycin-C, bleomycin dactinomycin, plicamycin, mitoxantrone, pixantrone, streptozocin); platinum dérivatives (e.g. cisplatin, oxaliplatin, carboplatin, lobaplatin, satraplatin); alkylating agents (e.g. estramustine, meclorethamine, melphalan, chlorambucil, busulphan, dacarbazine, cyclophosphamide, ifosfamide, hydroxyurea, temozolomide, nitrosoureas such as carmustine and lomustine, thiotepa); antimitotic agents (e.g. vinca alkaloids like Vinblastine, vindesine, vinorelbine, vinflunine and vincristine; and taxanes like paclitaxel, docetaxel and their formulations, larotaxel; simotaxel, and epothilones like ixabepilone, patupilone, ZKEPO); topoisomerase inhibitors (e.g. epipodophyllotoxins like etoposide and etopophos, teniposide, amsacrine, topotecan, irinotecan) and miscellaneous chemotherapeutics such as amifostine, anagrelide, interferone alpha, procarbazine, mitotane, and porfimer, bexarotene, celecoxib.
The efficacy of bispecific binding molecuie of the invention or polypeptides, and of compositions comprising the same, can be tested using any suitable in vitro assay, cell- based assay, in vivo assay and/or animal model known per se, or any combination thereof, depending on the spécifie disease or disorder of interest. Suitable assays and animal models will be clear to the skilled person, and for example include the assays described herein and used in the Examples below, e.g. a prolifération assay.
The data obtained in the experiments of the invention confirm that bispecific binding molécules of the invention hâve properties that are superior to those of binding molécules of the prior art. Among such properties are complété inhibition of the VEGF165-VEGFR2 interaction and a low IC50, as can e.g. be taken from the ELISA data of Figure 1 and Table 5 as well as the IC50 (nM) values for VHHs in the AlphaScreen assay as shown in Figures 3,17,18 and Table 7; and the affînity Ko (nM) of purified VHHs on recombinant human VEGF and mouse VEGF in Table 9, 10 and Figure 5. Also, as shown in Table 13, VEGF binders of the invention hâve high potency, i.e. in the subnanomolar range, in the HUVEC prolifération assay. This indicates that bispecific binding molécules of the invention are promising candidates to hâve therapeutic efficacy in diseases and disorders associated with VEGFmediated effects on angiogenesis, such as cancer.
According to another embodiment of the invention, there is provided a method of diagnosing a disease by
a) contacting a sample with a binding molécule of the invention as defined above, and
b) detecting binding of said binding molécule to said sample, and
c) comparing the binding detected in step (b) with a standard, wherein a différence in binding relative to said sample is diagnostic of a disease or disorder associated with VEGF- and/or Ang2-mediated effects on angiogenesis.
For this and other uses, it may be useful to further modify a bispecific binding molécule of the invention, such as by introduction of a functional group that is one part of a spécifie binding pair, such as the biotin-(strept)avidin binding pair. Such a functional group may be used to link the binding molécule of the invention to another protein, polypeptide or chemical compound that is bound to the other half of the binding pair, i.e. through formation of the binding pair. For example, a bispecific binding molécule of the invention may be conjugated to biotin, and linked to another protein, polypeptide, compound or carrier conjugated to avidin or streptavidin. For example, such a conjugated bispecific binding molécule of the invention may be used as a reporter, for example in a diagnostic System where a détectable signalproducing agent is conjugated to avidin or streptavidin.
Brief description of the Figures:
Figure 1: Purified monovalent VHHs block the hVEGF165/hVEGFR2-Fc interaction (ELISA)
Figure 2: Purified monovalent VHHs block the hVEGF165/hVEGFR1-Fc interaction (ELISA)
Figure 3: Purified monovalent VHHs block the hVEGF165/hVEGFR2-Fc interaction (AlphaScreen)
Figure 4: Purified monovalent VHHs block the hVEGF165/hVEGFR1-Fc interaction (AlphaScreen)
Figure 5: Binding of monovalent VHHs to recombinant human and mouse VEGF (ELISA)
Figure 6: Binding of monovalent VHHs to human VEGF121
Figure 7: Purified VHHs do not bind to VEGFB, VEGFC, VEGFD and PIGF
Figure 8: Formatted VHHs block hVEGF165/hVEGFR2-Fc interaction (ELISA)
Figure 9: Formatted VHHs block hVEGF165/hVEGFR1-Fc interaction (ELISA)
Figure 10: Formatted VHHs block hVEGF165/hVEGFR2-Fc interaction (AlphaScreen)
Figure 11:Formatted VHHs block hVEGF165/hVEGFR1-Fc interaction (AlphaScreen)
Figure 12: Formatted VHHs block mVEGF164/mVEGFR2-Fc interaction (AlphaScreen)
Figure 13: Formatted VHHs bind to mouse and human VEGF
Figure 14: Formatted VHHs do not bind to VEGFB, VEGFC, VEGFD and PIGF
Figure 15: Formatted VHHs bind to VEGF121
Figure 16; Sequence alignment of VHH VEGFBII23B04 with human VH3/JH germline consensus sequence
Figure 17: VHH variants of VEGFBII23B04 block hVEGF165/hVEGFR2-Fc interaction (AlphaScreen)
Figure 18: Sequence-optimized clones of VEGFBII23B04 block the hVEGF165/hVEGFR2-Fc interaction (AlphaScreen)
Figure 19: Sequence alignment of VHH VEGFBII5B05 with human VH3/JH germline consensus sequence
Figure 20: Description bivalent Ang2 VHHs
Figure 21: Purified bivalent Ang2 VHHs blocking hAng2-hTie2 (25-1), mAng2-mTie2 (25-2) and cAng2-cTie2 (25-3) interaction (ELISA)
Figure 22: Purified bivalent Ang2 VHHs blocking hAng1-hTie2 interaction (ELISA)
Figure 23: Description trivalent VEGFxAng2 bispecific VHHs
Figure 24: Purified trivalent VEGFxAng2 Nanobodies blocking hVEGF-hVEGFR2 interaction (AlphaScreen)
Figure 25: Purified trivalent VEGFxAng2 VHHs blocking hAng2-hTie2 interaction (ELISA)
Figure 26: Description trivalent and tetravalent VEGFxAng2 bispecific VHHs
Figure 27: Purified trivalent and tetravalent VEGFxAng2 VHHs blocking hVEGFhVEGFR2 (31-1) and hVEGF-hVEGFR1 (31-2) interaction (AlphaScreen)
Figure 28: Purified trivalent and tetravalent VEGFxAng2 VHHs blocking hAng2hTie2 (32-1 ), mAng2-mTie2 (32-2) and cAng2-cTie2 (32-3) interaction (ELISA)
Figure 29: Purified trivalent and tetravalent VEGFxAng2 VHHs blocking hAng2 mediated HUVEC survival
Figure 30: Description sequence optimized and affinity VEGFxAng2 bispecific VHHs
Figure 31: Purified VEGFANGBII00022-25-28 VEGFxAng2 VHHs blocking hVEGFhVEGFR2 (35-1) and hVEGF-hVEGFR1 (35-2) interaction (AlphaScreen)
Figure 32: Purified VEGFANGBII00022-25-28 VEGFxAng2 VHHs binding to human VEGF165 (36-1) and hVEGF121 (36-2) (ELISA)
Figure 33: Purified VEGFANGBII00022-25-28 VEGFxAng2 VHHs binding to (A) mouse and (B) rat VEGF164 (ELISA)
Figure 34: Purified VEGFANGBII00022-25-28 VEGFxAng2 VHHs binding to (A) human VEGF-B, (B) human VEGF-C, (C) human VEGF-D and (D) human PIGF (ELISA)
Figure 35: Purified VEGFANGBII00022-25-28 VEGFxAng2 VHHs blocking hAng2hTie2 (39-1 ), mAng2-mTie2 (39-2) and cAng2-cTie2 (39-3) interaction (ELISA)
Figure 36: Purified VEGFANGBII00022-25-28 VEGFxAng2 VHHs blocking hAnglhTie2 interaction (ELISA)
Figure 37: Purified VEGFANGBII00022-25-28 VEGFxAng2 VHHs blocking hAng2 mediated HUVEC survival
Materials and methods:
a) Production and functionality testing of VEGF109
A cDNA encoding the receptor binding domain of human vascular endothélial growth factor isoform VEGF165 (GenBank: AAM03108.1; AA residues 27 -135) is cloned into pET28a vector (Novagen, Madison, Wl) and overexpressed in E.coli (BL21 Star DE3) as a His-tagged insoluble protein. Expression is induced by addition of 1 mM IPTG and allowed to continue for 4 hours at 37’C. Cells are harvested by centrifugation and lysed by sonication of the cell pellet. Inclusion bodies are isolated by centrifugation. After a washing step with 1% Triton X 100 (Sigma-Aldrich), proteins are solubilized using 7.5M guanidine hydrochloride and refolded by consecutive rounds of overnight dialysis using buffers with decreasing urea concentrations from 6M till 0M. The refolded protein is purified by ion exchange chromatography using a MonoQ5/50GL (Amersham BioSciences) column followed by gel filtration with a Superdex75 10/300 GL column (Amersheim BioSciences). The purity and homogeneity of the protein is confirmed by SDS-PAGE and Westen blot. In addition, binding activity to VEGFR1, VEGFR2 and Bevacizumab is monitored by ELISA. To this end, 1 pg/mL of recombinant human VEGF109 is immobilized overnight at 4°C in a 96-well MaxiSorp plate (Nunc, Wiesbaden, Germany). Wells are blocked with a casein solution (1 %). Serial dilutions of VEGFR1, VEGFR2 or Bevacizumab are added to the VEGF109 coated plate and binding is detected using alkaline phosphatase (AP) conjugated goat anti-human IgG, Fc spécifie (Jackson Immuno Research Laboratories Inc., West Grave, PA, USA) and a subséquent enzymatic reaction in the presence of the substrate PNPP (p-nitrophenylphosphate) (SigmaAldrich). VEGF109 could bind to VEGFR1, VEGFR2 and Bevacizumab, indicating that the produced VEGF109 is active.
b) KLH conjugation of VEGF165 and functionality testing of KLH-conjugated VEGF165
Recombinant human VEGF165 (R&D Systems, Minneapolis, MN, USA) is conjugated to mariculture keyhole limpet hemocyanin (mcKLH) using the Imject
Immunogen EDC kit with mcKLH (Pierce, Rockford, IL, USA) according to the manufacturées instructions. Efficient conjugation of the polypeptide to mcKLH is confirmed by SDS-PAGE. Functionality of the conjugated protein is checked by ELISA: 2 pg/mL of KLH conjugated VEGF165 is immobilized overnight at 4°C in a 96s well MaxiSorp plate (Nunc, Wiesbaden, Germany). Wells are blocked with a casein solution (1%). Serial dilutions of VEGFR1 or VEGFR2 are added and binding is detected using a horseradish peroxidase (HRP)-conjugated goat anti-human IgG, Fc spécifie (Jackson Immuno Research Laboratories Inc., West Grave, PA, USA) and a subséquent enzymatic reaction in the presence of the substrate TMB (3,3’,5,5’w tetramentylbenzidine) (Pierce, Rockford, IL, USA). The KLH conjugated protein could still interact with VEGFR1, VEGFR2 and Bevacizumab, confirming that the relevant epitopes onVEGF165 are still accessible.
Example 1 is Immunization with different VEGF formats induces a humoral immune response in llama
1.11mmunizations
After approval of the Ethical Committee of the faculty of Veterinary Medicine (University Ghent, Belgium), 4 Hamas (designated No. 264, 265, 266, 267) are 20 immunized according to standard protocols with 6 intramuscular injections (100 or 50 pg/dose at weekly intervals) of recombinant human VEGF109. The first injection at day 0 is formulated in Complété Freund’s Adjuvant (Difco, Detroit, Ml, USA), while the subséquent injections are formulated in Incomplète Freund’s Adjuvant (Difco, Detroit, Ml, USA). In addition, four Hamas (designated No. 234, 235, 280 and 281) 25 are immunized according to the following protocol: 5 intramuscular injections with
KLH-conjugated human VEGH165 (100 or 50 pg/dose at biweekly intervals) followed by 4 intramuscular injections of human VEGF109 (first dose of 100 pg followed 2 weeks later with three 50 pg/dose at weekly interval).
1.2 Evaluation of VEGF-induced immune responses in llama
To monitor VEGF spécifie sérum titers, an ELISA assay is set up in which 2 pg/mL of recombinant human VEGF165 or VEGF109 is immobilized overnight at 4°C in a 96well MaxiSorp plate (Nunc, Wiesbaden, Germany). Wells are blocked with a casein solution (1%). After addition of sérum dilutions, bound total IgG is detected using horseradish peroxidase (HRP)-conjugated goat anti-llama immunoglobulin (Bethyl Laboratories Inc., Montgomery, TX, USA) and a subséquent enzymatic reaction in the presence of the substrate TMB (3,3',5,5'“tetramentylbenzidine) (Pierce, Rockford, IL, USA). For Hamas 264, 265, 266 and 267, an additional ELISA is performed in which the isotype-specific responses against VEGF165 and VEGF109 are evaluated. Isotype spécifie responses are detected using mouse mAbs specifically recognizing conventional llama lgG1 and the heavy-chain only llama lgG2 and lgG3 [Daley et al. (2005). Clin. Diagn. Lab. Imm. 12:380-386] followed by a rabbit anti-mouse-HRP conjugale (DAKO). ELISAs are developed using TMB as chromogenic substrate and absorbance is measured at 450nm. The sérum titers for each llama are depicted in Table 1.
Table 1: Antibody-mediated spécifie sérum response against VEGF165 and VEGF109
ELISA (recombinant protein solid phase coated)
Recombinant human EGF165 Recombinant human VEGF109
Llama Immunogen Total IgG lgG1 lgG2 lgG3 Total IgG lgG1 lgG2 lgG3
234 VEGF165-KLH + VEGF109 ++ n/d n/d n/d ++ n/d n/d n/d
235 VEGF165-KLH + VEGF109 ++ n/d n/d n/d ++ n/d n/d n/d
280 VEGF165-KLH + VEGF109 + n/d n/d n/d + n/d n/d n/d
281 VEGF165-KLH + VEGF109 + n/d n/d n/d + n/d n/d n/d
264 VEGF109 n/d ++ + + ++ ++ + +
265 VEGF109 n/d ++ + + + ++ + +
266 VEGF109 n/d ++ + +/- ++ ++ + +/-
267 VEGF109 n/d +/- +/- +/-
n/d, not determined
Example 2
Cloning of the heavy-chain only antibody fragment répertoires and préparation of phage
Following the final immunogen injection, immune tissues as the source of B-cells that produce the heavy-chain antibodies are collected from the immunized Hamas. Typically, two 150-ml blood samples, collected 4 and 8 days after the last antigen injection, and one lymph node biopsy, collected 4 days after the last antigen injection are collected per animal. From the blood samples, peripheral blood mononuclear cells (PBMCs) are prepared using Ficoll-Hypaque according to the manufacturées instructions (Amersham Biosciences, Piscataway, NJ, USA). From the PBMCs and the lymph node biopsy, total RNA is extracted, which is used as starting material for RT-PCR to amplify the VHH encoding DNA segments, as described in W02005/044858, For each immunized llama, a library is constructed by pooling the total RNA isolated from ail collected immune tissues of that animal. In short, the PCR-amplified VHH répertoire is cîoned via spécifie restriction sites into a vector designed to facilitate phage display of the VHH library. The vector is derived from pUC119 and contains the LacZ promoter, a M13 phage glll protein coding sequence, a résistance gene for ampicillin or carbeniciliin, a multiple cloning site and a hybrid glII-pelB leader sequence (pAXOSO). In frame with the VHH coding sequence, the vector encodes a C-terminal c-myc tag and a His6 tag. Phage are prepared according to standard protocols and stored after filter sterilization at 4°C for further use.
Example 3
Sélection of VEGF-specific VHHs via phage display
VHH phage libraries are used in different sélection strategies applying a multiplicity of sélection conditions. Variables include i) the VEGF protein format (rhVEGF165, rhVEGF109 or rmVEGF164), ii) the antigen présentation method (solid phase: directly coated or via a biotin-tag onto Neutravidin-coated plates; solution phase: incubation in solution followed by capturing on Neutravidin-coated plates), iii) the antigen concentration and iv) the elution method (trypsin or compétitive elution using
VEGFR2). Ail sélections are carried out in Maxisorp 96-well plates (Nunc, Wiesbaden, Germany).
Sélections are performed as follows: Phage libraries are incubated at RT with variable concentrations of VEGF antigen, either in solution or immobilized on a solid support. After 2hrs of incubation and extensive washing, bound phage are eluted. In case trypsin is used for phage elution, the protease activity is immediately neutralized by addition of 0.8 mM protease inhibitor AEBSF. Phage outputs that show enrichment over background are used to infect E. coli. Infected E. coli cells are either used to préparé phage for the next sélection round (phage rescue) or plated on agar plates (LB+amp+glucose2%) for analysis of individual VHH clones. In orderto screen a sélection output for spécifie binders, single colonies are picked from the agar plates and grown in 1 mL 96-deep-well plates. The lacZ-controlled VHH expression is induced by adding IPTG (0.1 -1mM final). Periplasmic extracts (in a volume of ~ 80 pL) are prepared according to standard methods.
Example 4
Identification of VEGF-binding and VEGF receptor-blocking VHHs
Periplasmic extracts are tested for binding to human VEGF165 by ELISA. In brief, 2 pg/mL of recombinant human VEGF165 is immobilized ovemight at 4“C in a 96well MaxiSorp plate (Nunc, Wiesbaden, Germany). Wells are blocked with a casein solution (1%). After addition of typically a 10-fold dilution of the periplasmic extracts, VHH binding is detected using a mouse anti-myc (Roche) and an anti-mouse-HRP conjugate (DAKO). Clones showing ELISA signais of >3-fold above background are considered as VEGF binding VHHs.
In addition, periplasmic extracts are screened in a human VEGF165/human VEGFR2 AlphaScreen assay (Amplified Luminescent Proximity Homogeneous Assay) to assess the blocking capacity of the VHHs. Human VEGF165 is biotinylated using Sulfo-NHS-LC-Biotin (Pierce, Rockford, IL, USA). Human VEGFR2/Fc chimera (R&D
Systems, Minneapolis, MN, USA) is captured using an anti-humanFc VHH which is coupled to accepter beads according to the manufacturées instructions (Perkin Elmer, Waltham, MA, US). To evaluate the neutralizing capacity of the VHHs, periplasmic extracts are diluted 1/25 in PBS buffer containing 0.03 % Tween 20 (Sigma-Aldrich) and preincubated with 0.4 nM biotinylated human VEGF165 for 15 minutes at room température (RT). To this mixture the accepter beads (10pg/ml) and 0.4 nM VEGFR2-huFc are added and further incubated for 1 hour at RT in the dark. Subsequently donor beads (10pg/ml) are added followed by incubation of 1 hour at RT in the dark. Fluorescence is measured by reading plates on the Envision Multi label Plate reader (Perkin Elmer, Waltham, MA, USA) using an excitation wavelength of 680 nm and an émission wavelength between 520 nm and 620nm. Periplasmic extract containing irrelevant VHH is used as négative control. Periplasmic extracts containing anti-VEGF165 VHHs which are able to decrease the fluorescence signal with more than 60 % relative to the signal of the négative control are identified as a hit. Ail hits identified in the AlphaScreen are confirmed in a compétition ELISA. To this end, 1 pg/mL of human VEGFR2 chimera (R&D Systems, Minneapolis, MN, USA) is coated in a 96-well MaxiSorp plate (Nunc, Wiesbaden, Germany). Fivefold dilutions of the periplasmic extracts are incubated in the presence of a fixed concentration (4nM) of biotinylated human VEGF165 in PBS buffer containing 0.1 % casein and 0.05 % Tween 20 (Sigma-Aldrich). Binding of these VHH/bio-VEGF165 complexes to the human VEGFR2 chimera coated plate is detected using horseradish peroxidase (HRP) conjugated extravidin (Sigma, St Louis, MO, USA). VHH sequence IDs and the corresponding AA sequences of VEGF-binding (nonreceptor-blocking) VHHs and inhibitory (receptor-blocking) VHHs are listed in Table 2 and Table 3, respectively.
Table 2: Sequence IDs and AA sequences of monovalent non-receptor-blocking anti-VEGF VHHs (FR, framework;
CDR, complementary determining région)
O Ol Ol Ol Ol Ol
H 0 EH 0 F 0 F 0 F 0 F 0
0 0 0 0 0 0 0 0 0 0 0 0
Ol > Ol > O > Ol > Ol > Ol >
z 0 F 0 Eh 0 F 0 F 0 F 0 F
Z 3 > 3 > B > B > B > B >
X rf X Z
Q 0
X eu 0 0 >4
3 0 Z X Q
O 0 F B >4
z Eu > rf Q
F Di H O X
Fi 0 0 rf Z
z 0 z 0 0
Z 0 0 Z X z
σι H eu z Z Z X
ai F H 0 z X Z >4 z
Q eu Z rf Z X Z 0 Z rf
u en 0 X Q Z Ol Z Z F
X X X X X X X X >4 >4 X X
> H d > > F > d 3 > > >
F rf rf rf F rf F rf F rf F rf
Z Eh 0 Eh Z 0 Z F Z F Z F
Z Q Z Q Z Q Z Q Z Q Z Q
Eh Q Q ω rf Z 0 Z rf Z rf Z
Z eu Z eu Z z Z Z Z Z z z
Q Eh Q z Q Z Q Z Q Z z z
Z Pi Fl Z z Z Z Z Z z z
0 0 0 En 0 0 0 0 0 0 0 0
M Z rf F Z rf F Z H Z F Z Z rf F Z rf
(H H S < Eh a rf F 3 Z F 3 rf F 3 rf
Ci tu O U Eu oi U Z Z O Z Ol U Z ot U Z Ot U
E z z x Z Z 0 Z Z X Z Z X Z Z >4 Z Z X
ω Eu X X >4 F
X Eh F >1 F F
Eh 0 F F o 0
Z Fl 0 Z 0 X 0 0 0 Q
0 0 0 Z 0 0 F Z 0 Z
X Pi 0 > 0 > 0 Z 0 > O >
cm z > 3 0 B 0 Cu > B 0 O 0
z ω ω Z Q e-1 Q F Z Z Q Z Z
Q F Q H F H rf M Q F 0 H rf
U rf 0 rf X rf X F B > F 0 X
Z Z Z F—i Z Z
0 0 0 0 0 0
CU 0 eu rf Z rf Z rf Z rf Z rf
0 > rf > rf > rf > rf r* rf >
ex E Ol Z Ol z Ol z OI z Ol z
Z M ni ω z z Z CH z z z z
ûj Z DÎ Z Ci z z >1 Z z z z z
E B Z B ω B Z B O B Z B Z
0 0 rf 0 0
rH s 3 > > 3
(4 0 Q P> F Q z
Q X 0 F H Q X
U 0 rf H Q F z
Z U Fl U Z U Z U Z 0 Z U
0 0 0 0 0 0 0 0 0 0 0 0
0 H 0 Fl 0 Z 0 Z 0 Z 0 z
0 Z 0 o ci ci 0 Z F 0 Z 0 0 Z 0 0 Z 0
0 Z Z 0 Fl Z 0 Z X 0 Z > 0 z z 0 z z
ω 0 0 ω 0 f Z Z F Z 0 rf Z 0 F z 0 0
>00 > Q Z >00 >00 > 0 Z > Q Z
Fl 0 0 FI 0 0 Z O F Z 0 0 Z 0 0 Z 0 0
H CH rf 0 CH rf F Ol rf 0 Ol z 0 Ol rf 0 CH rf 0
z > Ol rf > O rf > Ol rf > Ol rf > Ol rf > OI rf
E ω > Eh Z > > Z > F Z > rf Z > rf Z rf rf
n CO en O H CM n
X. § H 0 H 0 H 0 H CD H 0 F 0
Q 1—1 x. F X. M X. H X. H X. M \
h Q CÛ CM PQ Γ- 0 04 Z oo m cm m 0
tu O Z O Z F Z O Z O Z o
“ o 0 U 0 z 0 Q 0 0 0 m 0 m
tri pq 0 F Z F Z 0 Z 'M· Z 0 Z 0
> ω > o > O > O > O > o > o
ΟΙ Ol Ol Ol οι ο οι
en ω Eh en eh en h en μ ω eh en μ en
cd ω CD en CD en 0 U) 0 ω Ο en 0 en
< > en > Ol > Ol > ΟΙ > οι > οι >
CD μ CD Eh CD EH 0 μ 0 μ 0 μ CD μ
3 > 3 > s > s > 3 > 3 > 3 >
>4 X ω X
Q en ω Q
CO en μ X X
ta ea μ μ Ο
Eh μ μ μ
> > χ < Η
H H μ μ μ
eu >4 en en en < μ
z μ ea χ μ μ
eu CD 0 0 < μ
tz Eh ta Z X > μ
en ta μ μ 3 X en w
en >4 μ x ta x μ μ μ
q ta q ta en χ en Q
X X >4 >4 x x X X μ χ χ χ X
> > > > > Hi > H > > 3 > > >
μ < Eh < EH < H < ω < μ < μ <
Z Eh Z Eh Z en z en ζ μ bd ΕΗ Ζ μ
S Q S Q S Q Z Q Sa ο μ ο μ Q
tn ω < ω < ω < ω < ω μ ω Ζ Μ
z < Z 0j Z eu Z eu en μ ζ μ ζ μ
Q S q ta q ta Q μ q μ ο ο μ
μ μ ta μ «a μ ea μ μ μ μ μ μ μ
en en en en en en en co en en en en en en
H Z Eh > z < Ht z < μ z < χ z < Η Ζ XÇ > ζ <
Eh 3 Z Eh g Z μ s < H μ fi μ 3 « μ 3 < μ s <
ta O U ta Ol U ta οι u μ οι u μ οι cd μ ο cd μ οι cd
μ μ >4 ta μ >4 ta μ x ea μ x μ μ χ μ μ χ μ μ χ
>4 >4 x x ζ ω Η
< Eh CO μ μ μ μ
Eh [ï4 en μ Ζ μ en
Eh CD CD eu CD μ 0 U3 0 en 0 Η4 0
CD CD cd ta < ta < μ 0 μ 0 μ μ μ
CD Z CD > en > C0 > μ > 0 > ζ >
en > en en 3 co 3 co ο< en μ en 3 en
en en en Q μ Q μ Q ζ ο en ω en ο
H Q h μι 1-4 < Η < Η < Ηι < n μ
dû > ta >h < X < X C χ ζ χ μ X
« ta ta bd μ μ μ
CD CD CD 0 0 0 ο
P4 < eu < eu < μ < μ en eu <
< > < > < > < > < > < >
O μ οι μι οι Eu οι μ οι μ οι μ οι μ
μ ω ta ω ea ta ta rf μ ω μ w μ ω
x ce; >4 ta μ en μ ta μ μ μ μ μ μ
3 z 3 Ol 3 UJ 3 μ 3 μ 3 ω 3 ω
ο ο
§ s â Σ 3 3
5* 5> 5J > X
s? z μ en X Ζ X
en z H Η ζ ο en
μ u μι o μ o μ ο μ cd μ ο μ cd
cd en cd en cd en 0 C0 0 en 0 en 0 en
cd μι cd μ cd μ 0 μ 0 μ 0 μ 0 μ
CD Ci S cd ta en cd ea en ο μ en 0 ea en 0 μ en 0 μ en
en μ E en μ ta en μ x en μ χ en μ μ en μ μ en μ μ
ω en μ w en h [il CO H ω en η ω en μ ω en ω ω en μ
> CD H > CD z > 0 en > 0 CQ > 0 μ > ο μ > 0 μ
μ CD CD μ o cd μ 0 h μ 0 μ μ 0 ο μ ο ο μ ο ο
o» eu en oi eu en Oi < en οι < en Ο μ en Οΐ < en οι < en
> ot < > oi <; > Ol < > οι > > οι > Ο < > οι μ
ω > > ω > < ω > < ω > < ω > <! ω > fi ω < μ
m eo X ω σ> ο
H VO h eo m to Η tO Η Ό Μ tO Η X
H X. H x. M X, Η X. Η X. Μ X. Ht X.
en in en exi en ω m ό μ co Ζ σι μ χ
[U o μ o μ o μ ο μ ο μ ο μ ο
CD ffl CD CD O < 0 < 0 ο 0 Q ο ω
Μ LT) ω co ω x ω χ ω χ ω œ ω œ
> O > o > o > ο > ο > ο > ο
fa CM EH CM CM CM CM
H 0 Eh en Eh 0 Eh 0 Eh 0 H 0 Eh 0
0 ω 0 en 0 0 0 0 0 0 0 0 0 0
ο > CM > < > CM > fa > CM > O< >
ο ε-< 0 Eh 0 En 0 Eh 0 Eh 0 Eh 0 Eh
s > S > S > B > B > B > B >
< fa X fa fa
X 0 fa 0 0
en H X EH Q
0 X B X EH
fa B fa B X
X Q a Eh
X 0 H o X
X fa a 0 a 0
ω Eh 2 0 fa o Q
Μ 0 fa fa 0 fa fa X 0
0 0 fa fa Q H fa 2 2 X
ω B 0 fa X Eh Pi X B 2
fa CM 0 0 fa fa 0 fa X X
fa fa X fa fa Eh 0 fa EH fa B
X X 2 X X X X X X X X X X X
> > > > > > S > > > s > > >
Η < EH < EH < Eh < En < En a in a
2 η •Z. Eh g eh 2 Eh 0 Eh 2 En 2 eh
a ο fa Q S Q fa Q fa Q fa Q fa Q
a ω < fa 0 fa < fa 0 fa a fa a ω
Q fa 2 fa 2 a 2 fa 2 0 2 fa 2 fa
Q fa Q fa Q fa Q fa Q fa Q fa Q fa
fa fa fa fa fa fa fa fa fa fa fa fa fa fa
0 2 CO 0 0 0 0 0 0 0 0 0 0 0
H 2 a H g 0 H 2 Eh fa 2 < h 2 a fa 2 a h 2 a
Eh S 2 EH S < eh S 2 eh s a eh s a Eh S 3 eh s a
fa CM U fa CM 0 fa CM 0 fa cm 0 fa CM 0 fa CM 0 fa OI U
fa fa X fa fa X fa fa X fa fa X fa fa X fa fa X fa fa X
X CM X X Eh X X
X EH < Eh X EH En
H Q EH 0 > 0 2
CO 0 0 Eh 0 0 2 0 0 0 0
0 0 < fa 0 0 EH < Q 0 Eh fa fa fa
fa fa en > fa fa 0 > X fa 0 > 0 >
CO > B en 0 > B 0 fa > B 0 fa 0
fa co EH Q 0 0 fa Q 0 0 fa Q Eh Q
H 0 H < H Q H 0 H Q H 0 H Eh
fa < < X fa > > Eh < a > Eh > X
fa fa fa fa ta fa fa
0 0 0 0 0 0 0
fa < fa 0 fa < fa < fa H fa a fa a
< > < fa < > a > a > a > > >
CM fa CM fa CM fa OI fa CM fa CM fa CM fa
fa fa fa fa fa fa fa fa fa fa fa fa fa fa
X fa fa fa X fa fa fa fa fa fa fa fa fa
B O s ω B æ B fa B fa B fa B fa
0 0 0 0 0 0
> > S
p> Æ rf Q 0 Q Æ
Q X 2 Q X Q X
CO fa 0 Eh fa Eh 2
fa 0 fa 0 fa 0 fa U fa U fa U fa O
0 CO 0 0 0 0 0 0 0 0 0 0 0 0
0 fa 0 a 0 fa 0 fa 0 fa 0 fa O fa
0 fa fa 0 fa 0 0 fa 2 0 fa 0 0 fa 0 0 fa 0 0 0 0
CO fa > en fa fa 0 fa fa 0 fa fa 0 fa fa 0 fa fa 0 fa fa
fa CO IH fa 0 Eh fa 0 fa fa 0 Eh fa 0 0 fa 0 Eh ω 0 a
>00 >00 > 0 H > 0 fa > Cl fa > 0 fa > ο a
fa 0 0 fa 0 0 fa 0 0 fa 0 0 fa 0 0 fa 0 0 fa 0 a
Oi fa co CM a 0 CM fa 0 CM a 0 CM fa 0 oi a 0 cm a 0
> CM à > O > > O < > CM < > o a > o a > cm a
fa > a fa > < fa > > ω > a ω > a ω > a ω > a
r-1 0 H* 0 ko x
H X h r* IH X H X IH X H X H X
H X H X H X H X IH X H X H —
en co en x CQ σι CQ CM CQ 0 en 0 en x
fa o fa o fa o ta i—1 fa O fa O fa o
0 fa 0 fa 0 a 0 < 0 Q 0 fa 0 o
fa œ fa œ fa 0 fa 0 fa 0 H en fa o
> o > o > O > o > o > o > rH
Ο Oi ot ot OI OI oi
Εη X EH en Eh en Eh en X X En en Eh X
ο x 0 en 0 en 0 en 0 Ui 0 U) 0 X
ο > o > ot > ot > a > Ot > Ot >
0 Η 0 Eh 0 EH 0 En 0 Eh 0 Eh 0 Eh
5 > & > Z > Z > Z > Z > Z >
χ X en en X X X
X fi en Eh *=5 X
Ζ 2 Eu XI X X X
Q eu 0 t-q X
ai M Eh Q Q Q x
z X z X X CU
2 en eu Q Z Z X
ο Q Q en Q Q 0
X en Z z X X X
X 0 Q X X X X
S X te z X en X
Z Ém Eï-i X X Eh Eh tu
fi X X X > z Eh X Eh X X
X X Q ci S X x ex O O 0 Q
X X X X X X X X X X X X Z X
> > 0 eh > X > h > rf > < > >
Eh < Eh < Eh < Eh < Eh < Eh fi EH <
Z Eh Z Eh Z Eh Z EH Z Eh Z Eh Z Eh
2 Q X Q X Q 2 Q X Q X Q X Q
< x < M < ω < ω < X < X < X
z eu Z X z eu Z x Z x Z X Z X
Q Z Q X Q 2 q ω 0 X 0 X Q X
X X ai X ps x > XI X X X X X X
x ω en en en en en en en co en en X CO
H z < > Z fi H Z X > Z < H Z < Ht Z H z <
E S i < s Eh S < x s fi Eh S fï Eh S fi X S fi
X Ot U Eu O U X ΟΙ O x 2 o X Ot U fa a u X ot 0
X X x XXX ci X en X XI X XXX XXX XXX
X en X X X X X
Eh Eh Eh Eh Eh Eh Eh
z Ci Q en Z X
H 0 X 0 X 0 en o < 0 < 0 X 0
0 X 0 2 0 X 0 X 0 X 0 X 0 X
en > en s X > en > en > X > X >
en < Eh en x en Z en Z en Z en Ot X
en Q Z Q x en 0 Q en Q en Q X Q
H f£ M < M < Ht < Ht < Ht < Ht <
Q > en x X X < X X X X X EH x
2 X X X X X X
0 0 0 0 0 0 0
eu < eu < eu < X < X < x fi X <
< > < > < > < > < > < > fi >
o x Ot & Ot x CH X ot X oi X OI x
X > X M x ω X X x x x ω X X
X a x pi X X X X X X X X X X
Z K) z x z ω z X Z X Z X z x
0 0 0 0 0 0 0
2 s X Ht H S
S ;s z 0 < 2
X X X X X X X
z Ci z X ω en X
x u X U X U X 0 X 0 x o X o
0 en 0 en 0 en 0 en 0 en 0 tn 0 X
0 X 0 X 0 Z] 0 x 0 X 0 X 0 X
0 a en c K en 0 X en 0 x en 0 s en 0 S en 0 X X
en x in W J hl en ni fa en x x en x x en x x XXX
W en Eh M en EH ω en eh X en eh x en X en < X X EH
> 0 ai > 0 Q > ω h > 0 X > 0 x > 0 X > 0 X
X 0 0 X 0 O x ο ο X 0 0 X 0 0 X 0 0 X 0 O
O < en Oi < en en < en Ot < en ot < en ot fi en CH < X
> o < > O rtj > ot < > ot <; > ot fi x oi fi > Ot <
x t> < ω > «J x > > X > fi x > fi x > < x > fi
00 en O rH CM en H*
Ht t· Ht X H 00 H 00 Ht 00 Ht 00 Ht 00
H X. Ht Hl \ H 'x. H \ Ht \ Ht X.
ff) r- en X m m œ X <-H X OT X
X O Cu O X o X o X rH X O X o
0 w 0 0 0 0 0 o 0 O 0 Q 0 X
x ο x o ω o X X X X X X X X
> f-H > rH > r-t î> T—1 > T—1 > 1—1 > i-l
Ο Oi OI OI O OI A
A ω Eh 0 A 0 A 0 A 0 A 0 A 0
ο 0 0 0 0 0 0 0 0 0 0 0 0 0
οι > Oi > oi > oi > OI > A > OI >
0 Εη 0 EH 0 En 0 A 0 A 0 A 0 A
Β > B > B > B > S > A > B >
X X X X A
a Q O Q A
ο 0 > 0 A
ω od eu cd A
en En Cd A X
X > En > 0
X H A H 0 A
A 0 0 X A X
0 Dd Eh cd A X X 0
Η 0 0 0 X A Z A
X A X bd A X A 0
A Pu £n tu Q A < 0
od x od X > X od X M Q 0 A
ο ο Q Qd 0 Cd Q Cd Q > 0 A
Pu X X X X X X X A X X X X X
A Μ > H > > > H H > A d > >
Η < EH < En < A < bd < A rt] A <
β η Z 0 Z Eh Z 0 Z A Z A Z A
bd Q A Q bd Q A es A ο Z Q Æ Q
< a <! a < a < a A A < A PÜ A
Z A Z eu 0 eu Z eu Z A Z A Z A
Q A Q A Q bd Q bd Q 3 Q A Q A
A A A A Cd A Dd A 0 A A A A A
ω ω 0 0 0 A 0 0 0 0 0 0 0 Z
Η Ζ > h z H g Eh H Z < H Z H > Z bd H Z <
A 3 < Eh > < EH g < A 3 3 A 3 < 0 3 < A 3 Z
Pu ΟΙ Ο Pu OI Q q oi u tu oi U A ΟΙ O A OI U A Oi (J
ai q χ A A X A A X Dd A X cd A x A A 0 A A X
X Sx 0 X X X X
A 0 A A A A X
A 0 A A eu A
0 ο DU 0 l-H 0 Du 0 0 0 A 0 0
0 A < A 0 bd < bd a bd 0 A 0 0
0 > 0 > 0 > 0 > A > 0 > A A
B X B 0 oi 0 B 0 0 0 A 0 0 >
5d Q En Q 0 Q A Q A C3 0 Q A 0
Pu < H < 1-H < H < H < H > H 0
< X < X 0 X < 0 < X A X A <
A bd bd bd 2 A A
0 0 0 0 0 0 0
DU < eu < Ou < eu < A < A 0 A <
< > < > > > < > < > < > < >
O A oi Pu Oi tu OI L· Oi tu Oi B Oi A
A A Dd M A a Dd A Dd tl A A A A
Pu Dd tu Dd tu cd bu Cd A Dd > A X A
3 M B t] B Q B a B Q B 0 B Oi
0 0 0 X 0
s S S 3
Sj B *>
en EH A A X 0 O
Pu H 0 M bd 0 0
A U a u A O A U A U a y A U
0 0 0 0 0 0 0 0 0 0 0 < 0 0
0 J 0 »-1 0 A 0 A 0 A 0 A 0 A
0 Dd en 0 Dd 0 0 Dd 0 0 Cd 0 0 Cd Z 0 A 0 0 A A
to q Pu 0 A X 0 A tu 0 A X 0 A A 0 A A 0 A >
a 0 a M 0 H A 0 0 A 0 H A 0 A A 0 A A 0 H
> 0 A >00 > O cd >00 > 0 A > 0 A >00
A 0 0 •-1 0 EH A 0 0 A 0 A A 0 0 A 0 0 A 0 0
O< a 0 ex < en OI 0 0 Oi < 0 Oi < A Oi A 0 OI A 0
> O m > Oi rf > Oi < > Oi < > Oi > > a < > oi <
A > F< M > i< M > A M > < a > 0 A > < A >
in Φ r* 00 en O H
H 00 H 00 H 00 H 00 H 00 h en H 01
H \ H \ H 1-H \ W \ M \ H \
OQ m OQ O CQ CXI m «r. m r- A I-H A 'O’
tu o tu rH tu rH A O A o A o A O
0 A 0 tu 0 tu 0 0 0 < 0 A 0 U
M ï—1 A rH M <-l A A a cn A (XI A CM
[> i—1 > rH > rH î> << > <-H > M î> i—l
CH O OI OI O Oi CX
Eh 0 Eh en Eh 0 EH 0 EH 0 Eh 0 Eh 0
0 0 0 en 0 0 0 0 0 0 0 0 0 0
οι > Oi > OI > Z > OI > O< > Oi >
0 &4 0 Eh 0 Eh 0 Eh 0 Eh O Eh 0 Eh
s > S > 3 > S > B > B > B >
0 < 0 X 0 0 0
EH >l 0 0 Eu Eh Eh
X 0 Eh 0 >4 >4 >4
Z 0 >4 0 0 >4 >4
X Z B Q 0 0 0
0 >4 >4 B B B
0 >4 0 Q 0 0 0
X >4 < 0 0 0 0
Z >4 Eh 0 0 X >4 >4 0 X X
X K 0 0 >4 0 Eh Q < 0 0 Q
0 x 0 0 Z B 0 >4 0 >4 0 X
Q 0 3 OÙ >4 H H 0 Q 0 Q 0 o
0 < Oi 0 X 0 Q 0 Z 0 Z 0 Z
Q > OÙ B 0 Z 0 >4 Q > Q > Q >
Cm >4 Z >4 >4 >4 >4 X >4 >4 >4 X X X
> > > > S > > > > > > > > >
0 < Eh < Eh < Fc < Eh < Eh < H <
Z En Z t4 Z Eh Z Eh S Eh 0 Eh Z Eh
0 Q Où Q £ Q £ Q £ Q £ Q S Q
F· 0 < 0 < 0 < 0 < 0 < 0 < 0
Z eu Z eu Z 0 Z 0 Z 0 Z 0 Z 0
Q OÙ Q 0 Q 0 Q Eh Q £ Q £ Q £
0 0 i·^ 0 0 0 0 0 0 0 0 0 0
en en 0 0 0 0 0 Z 0 0 0 0 0 0
H Q H H Z 0 0 Z < H Z 0 H Z > H Z > H Z >
H S < Eh S < Eh S < Eh S < Eh S < E4 S < Eh S <
0 O U 0 CX U 0 Oi U 0 Oi U 0 o< U 0 OI U 0 Ol U
θΰ t—l 0 0 X Où 0 >4 0 0 X 0 0 X 0 0 X 0 0 X
>4 OI >4 X >4 >4 X
EH EH EH Eh Eh Eh Eh
eu Q 0 0 S Ξ S
0 0 0 0 0 0 0 0 0 0 0 0 0 O
Z 0 < 0 Eh 0 0 0 Z 0 0 Ol 0 0
Eh > 0 > 0 > 0 > Eh > 0 > 0 >
en w B 0 B 0 B 0 B 0 B 0 B 0
En Q Eh Q 0 Q Eh Q en Q 0 Q 0 Q
H < H < H 0 1—1 eu M < H < M <
< >4 < >4 > Eh < 0 > X > >4 > X
Z 0 0 0 0 0
0 0 0 0 O 0 0
eu < eu 0 0 < eu Eh 0 < 0 < 0 <
< > < > < > < r» < t* < > < >
ex L· oi 0 CH Cm O< 0 Oi 0 Oi 0 Oi 0
ce ω où ω 0 0 0 0 0 0 0 0 0 0
Cm OÙ 0 S 0 0 0 0 0 0 0 0 0 0
β 0 B 0 B 0 B 0 B 0 B 0 B 0
0 0 0 0 0 0 0
> 2 S S S
£> Q Q Q Q Q
>4 >4 Q >4 >4 X X
Z 0 Eh 0
0 U 0 0 0 0 0 U 0 O 0 U 0 0
0 0 0 0 0 0 0 0 0 0 0 0 0 0
0 0 0 0 0 0 0 0 0 0 0 0 0 0
0 Où z 0 OÙ 0 0 0 0 0 0 0 O 0 0 0 0 0 0 0 0
000 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0
0 0 Eh W 0 EH 0 0 EH 0 0 Eh 0 0 Eh 0 0 EH 0 0 H
>00 > Q Z >00 >00 >00 >00 >00
000 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0
Oi < 0 OI 0 0 OI < 0 OI < 0 Oi < 0 OI En 0 Oi < 0
> O < > OI > > OI fi > Oi < > Oi < > Oi < > Ol <
M < Eh 0 > < 0 > ri 0 > < 0 > < 0 > < 0 > <!
CM 0 0 CO r- 00
H CS H <S H CS H CS t-4 es w es 1—1 CS
H \ H X. H X H X H X H X» 1—1 X.
CO O 0 0 0 0 <-H 0 œ 0 0 CQ kD
Cm t-4 0 O 0 O 0 tH 0 o 0 O 0 O
0 0 0 0 0 U 0 0 0 u 0 0 0 <
0 CM M cm 0 CD 0 CD 0 CD 0 x 0 CO
> rH > rU î> <”< > M > 0 > 0 > 0
οι Ol Ol O> Ol Ol Q*
η ω EH 0 Eh 0 Eh 0 Z 0 Z 0 a 0
ο ω 0 0 0 0 0 0 0 0 0 0 0 0
ΟΙ > Ol > Ol > Ol > Ol > Ol > Z >
Ο Ζ 0 Eh 0 Eh 0 Z 0 H 0 Z 0 Z
Β > S > Z > z > B > B > B >
ο 0 X 0 0 Z Z
Ζ En X EH Z 0 a
X X X X X a X
X X z X X 0 U
Ζ z Z z z z
Β Z z Z B > X
αύ z Z Z O a
en 0 z 0 0 Q 0
X X X X 0 X X X X 0 U
ζ ζ Eh Z z Eh Z a q 0 X X
Ζ X Dû X a Z X Z X X Q Q
Ζ Ζ DÛ Z Ht z z z z X > X
ζ ζ Dû Z Z z z Z Z Z X Z Z
ο > Q > 0 Q > 0 > 0 z Z Z
0 χ 0 X X X 0 X z x X X Z x
> > > > ? > > > Ol > > > z >
Ζ < Eh a eh a Eh a z a z a z a
Ζ Ζ Z Eh Z eh z Z z z z z z z
Ε ο Z Q Z Q g Q z o Z o o» o
a ζ a w a z a z a z a z a z
E ζ Z CM z z Z z z z z z z Z
q ζ Q Z z z Q Z z z Q Z o a
Dû Ζ Z Z z z z z z z Z Z z z
en co 0 0 0 0 0 0 0 0 0 0 0 z
Ht Z > h z a n z a Ht Z > H Z > x z a ni z a
Ζ S a Eh S a Eh S a EH s a z s a z z a h s a
fa O U Z Ol O Z Ol U zou Z oi Q Z oi U z OI o
Z Z X Z Z X Z Z 0 z z X Z Z X Z Z X Z Z X
X X 0 X Q X X
Eh Eh Eh Eh Z z z
S S Z S s 0 s
0 0 Q 0 Q 0 0 0 o 0 0 0 0 0
0 Z 0 Z 0 Z 0 z 0 z 0 z 0 z
Eh > Eh > Eh > z > 0 > 0 > 0 >
s en Z 0 Z 0 B 0 B 0 Z 0 B 0
0 Q 0 Q 0 z 0 Q 0 Q Z Q 0 Q
H < h a Ht a Ht < ni a Ht < Ht Z
> X > X Eh X > X > X Z X a x
z z Z Z z z z
0 0 0 0 0 0 0
eu < z a z a z a z a z a Z 0
a > a > a > a > a > a > a n
O Z Ol z Ol z Ol z Ol z ot 0 oi z
z z z z z z z z Z Z z z z z
Z Z z z z z z z Z Z z z z z
b z z z s z B Z B Z B z s z
o 0 0 0 0 0 rf
s s S Σ S s S
Q Q Eh Q Q rtj rtj
X X X X X X B
a a > a a 0 Z
z υ z U Z U Z U z o Z U Z U
0 en O 0 0 0 0 0 0 0 0 0 0 0
0 μι 0 Z 0 Z 0 Z 0 Z 0 Z 0 Z
0 Z 0 0 Z 0 0 Z 0 0 Z 0 0 Z 0 0 Z Z 0 Z Q
0 Z Z 0 Z Z 0 Z z 0 Z z 0 Z Z 0 Z z 0 Z z
Z 0 Z M 0 Eh Z 0 Eh Z 0 Z Z 0 Z Z 0 Z Z 0 Z
> 0 Dû > 0 Z > 0 Z S 0 Z > 0 Z > Q Z > Q Z
Z 0 0 μι 0 0 Z 0 O Z O 0 Z 0 0 Z 0 0 z 0 a
Ol < 0 οι a 0 oi a 0 oi a 0 oi a 0 o< a 0 oi a 0
> οι a > oi a > o a > oi a > oi a > o a > oi z
z > a z > a z > a z > a z > a z > z z > z
en
H Ct H Ht I—I Ht Ht Ht
Ht X. M ·χ Ht X. Ht X. Ht X, Ht X. Ht x»
CQ rH CQ eo Z 0 0 <—I m o 0 rH 0 rH
Z rH Z o Z O Z rH Z rH Z rH Z t—t
0 Z 0 « O 0 O H 0 Q CM 0 Z 0 0 U 0 Z 0
M en Z rH O Z rH O Z rH O Z CM O Z Φ O Z CD O
> m > ·=3* H > rH > 'T H > 'T rH > 00 rH > 00 rH
Ο Ol Ol Ol O Z O<
E-· en tu en eh en eh en H en Z en eh en
ο ω 0 en 0 en 0 en 0 en 0 en 0 en
οι > Oî > z > Oi > Ot > Z > Ot >
Ο Η 0 Eh 0 Eh 0 Eh O Eh 0 H 0 H
Β > B > B > S > Z > S > B >
Ε-Ι ω X X z 3
rt z Z Q 3 0
2 en X X 0 Z
Η ω en 0 U
X frj 0 Z > Z
Ζ H X 0 H
en Z z b> Q
Η en en X j> Z
ω Pi en z Eh X 0
X eu πΊ Q Z Eh Z 0
X X < 0 en 0 tn Q O
en Q en x z Ol < Z O Z
Du X EH PU en < en Z Eh 0-j
q ζ Ol X Eh 0 Q Eh X Q 0
< χ X X Q X X X Z X X X X X
> > > > > > 0 Z z t> > > > X
Ε-< < Eh < H < < < Eh < Eh < Eh <
Ζ Εη Z Eu Z Eh Z Eh 2 Eh B En 3 Eh
Ζ Ο Z Q Z Q Z O Z Q Z Q Z Q
ο ζ > ω < ω rf Z < Z < Z < Z
Ζ Ζ X PU Z H Z Z 3 en Z Z 3 Z
Q Ζ Q Z Q Z Q Z O Z Q Z Q Z
Ζ Ζ Z t-3 Z Z Z Z Z Z Z Z tn z
co ω en en en en en en en en en en en Q
Η ζ < H Z < hT ΪΣ 3 Z < H z z X 3 < x 3 >
Η 3 ο H S < Eh S < h S < Eh Σ < X 3 < En X <
α ο Z oi O Z Ol O Z Ol U z οι u Z Ol O z οι o
ci Ζ X Z Z X Z Z X Z i-3 X Z Z X Z Z X z z x
X X X Cm 3 X X
X Eh z Eh z EH
Εη M z en Cm 0 Eh
Εη Z 0 en 0 0 0 Z 0 0 0 0 0
0 Ο > z 0 z X z 0 z 0 z > Z
en ζ Eh > en > En > > > tn > Q >
ζ > B en Z en B en z en z en n tn
Eh < Z Q en Q tn Q z z H O H Q
Μ Μ X < H > H < M < H < X <
< < < X X X en x en x < X O X
Ζ EH z Z Z z z
0 0 0 0 0 0 0
Pu < Pu < Pu < z < z en Z < z en
0 > Eh > < > Eh > < > < > < >
οι ζ O< z oi Eu Z Z ΟΙ B Ol z oi 0
ci ζ z ω z ω z z Z z z z z z
Ζ Ζ z z z z Z Ci & z z z z
Β ζ s ω B ω B Z 3 0 B Z B Z
ο 0 0 0 Z Q 0
3 S > s S S X
ζ Eh Eh ΕΞ B 5j Q
Η X X X en X X
en Eh Z en EH z Q
ζ ο Z U Z U z o Z o z o Z O
ο en 0 en 0 en 0 en o en 0 tn 0 en
0 1-3 0 1-3 0 z 0 z 0 z 0 z 0 z
U Pi EH 0 Z Z 0 z en 0 z z 0 z en 0 z tn 0 Z 0
en z z en >-3 z en z z en z z en z z tn z z en z z
z en z ω en h ω en eh z en < z en eh z en Eh z en x
> Ci Z > 0 Z > 0 z > 0 en > 0 Z > 0 Z > 0 z
Z 0 0 Z 0 0 Z 0 0 Z 0 0 Z 0 0 Z 0 Z Z 0 0
oi h en oi < en o < en oi < en oi z en O < en O < en
> o <; > Oi < > o > O < > Ol < > Ol < > oi
z 3 < z > < z > < Z > EH Z > EH z > < z > <
H M n h-i HH H 1—1
H X X X X X X X M X H X X X
m œ m o m x m œi m x CQ r-i Z exi
Z o Z X Z i—1 Z o z O Z o Z o
0 0 40 0 0 X 0 0 œ 0 Z (Λ 0 z o 0 < H 0 C CM
Z <O O z *x> o z o Z *Ό O Z X H Z 00 H Z 00 H
> 00 X > CO r-l > 00 r-l > 00 H > 00 H > 00 H > 00 r-l
A CM CM CM CM CM A
A CO Eh 0 E-< C0 A 0 H 0 H 0 A 0
0 CO 0 0 0 0 0 0 0 0 0 0 0 0
X > O > CM > CM > O > O > X >
0 EH 0 EH 0 A 0 A 0 A 0 A 0 H
co > S > S > B > S > B > B >
x EH X 3 0 X
a X X 3 X X
X X fi {X Q 0
x x 0 EH a < X
w pi oi X
M S A Æ X X
Pi 0 x 52 CM U X
Pi X X HH X X X >
Pi X 0 HH X X Q Q
A 0 0 Z X 0 0 0
O A O X 0 X X X B
x x Pi 0 X 0 X H Q X X
Ρί A Dm X Q X Q 0 X X X B
x A < Q X X X X 0 < X X
X >1 X X X X X X X X X X X X
A > > > S > 0 > > > æ > 5:
Eh 0 EH < EH < H 0 A < A < A <
Z Eh Z EH Z EH Eh Eh Z A Z ÈÎ jZ Eh
X O X Q S Q X Q X Q X Q X Q
< x < X < x Q X < X < X SJ X
CO H Z X z X Z x Z X Z x Z x
Q 0 Q X Q X Z X Q X Q X Q X
pî a Ci A X A HH A X A A A X A
0 0 0 0 0 0 0 > 0 0 0 0 0 0
H Z Æ Cm Z < HH Z < H z < HH Z < H JS < 1—1 H rîî
A S fi Eh S fi A S < 0 s < H S fi H S fi H 3 S
Cm O 0 X Oi U X CM 0 X CM 0 X CM O X CM U X CM O
x a x Pi A X X A X X A x X A X X A X X A X
X Q X X X B X
Eh H Eh H A X Z
> 0 A 0 HH HH A
0 0 Eh 0 < Q S 0 0 0 0 0 HH
Q X a X 0 X X x 0 (X Q X 0 0
X > 0 > 0 > 0 > 0 > 0 > 0 X
0 X 0 0 B < 0 0 A 0 < 0 0 >
0 Q 0 Z A Q A Q X Q 0 < A 0
M < HH A HH < HH < HH X S A HH Q
U X O X X X < X < X U X A >
X x X X X X X
0 0 0 0 0 0 0
Oh co X 0 X < X < X < X 0 X
< > < > < > < > 0 > < > < >
CM 0 o < CM X CM X CM HH Oi < CM A
Pi x Pi M x ω X X X X X X X X
Cm Pi X X X x x x X x X X X X
& x & x B X g x B X B X B CM
0 0 0 0 0 0 0
M H > 3 S H 2
< < Z Q X
X X A X 0 X Z
Q Q 0 A 0 A A
A 0 A U A U A 0 A U A U A U
0 0 0 0 0 0 0 0 0 0 0 0 0 0
0 A 0 A 0 A 0 A 0 A 0 A 0 A
0 Pi Q 0 X Q 0 Pi 0 0 X A 0X0 0X0 0X0
0 J A 0 A A 0 A X 0 A X 0 A X 0 A A 0 A 0
x 0 z x 0 x ω co A X 0 0 X 0 H X 0 H X 0 HH
> 0 J > 0 X >00 > < Q > 0 X > 0 X >00
Hl 0 0 A 0 0 A 0 0 A 0 0 A 0 0 A 0 0 A 0 H
O CM 0 O» X 0 CM < 0 CM X 0 CM < 0 CM < 0 CM < 0
> O < > CM < > CM rf > CM < > CM < > a < > CM Æ
X > En X > > ω > < X > rtj x < rf| x > > X > <
H HH HH HH HH HH HH
HH H \ HH X. HH -V. HH X. HH X. HH \
m cn X OJ 0 0 X 0 X rH 0 rH 0 M·
x o X O X O X O X rH X O X O
0 m ω 0 ω 0 0 0 0 0 0 0 0 t 0 æ œ 0 0 σι
ω co h X CO rH X 00 rH M 00 H X 00 H X 00 H x σι H
> oo H > 00 H > 00 H > 0 H > 00 H > 00 rH > oo H
Ο οι Ol Ol Oi
ζ ω Eh 0 Eh en Eh en Z 0
Ο 0 U 0 0 0 0 en 0 0
Ο > Ο > Oi > Oi > Ol >
Ο Ζ 0 Εη 0 Eh 0 Eh 0 Z
3 > 3 > 3 > 3 > 3 >
eu Pd Q
X 0 en X X
Ο Εη x Q Z
*25 Ζ z P Cd
*> Pu X S Ol
[> ζ en J> Q
0 > en en cd
X ζ X X 0
Ζ ηΰ Z » Z 3
ω ω X Z fa z
PU 0 0 Z X fa 0
Ρΰ > υ Q Z Pd 0
Η cd fa fa Q H z
< Q Q Q > < Z X
0 X X X fa x en x Z X
> > > > > > > £ X X
Ζ < Eh < En < Eh < £ M
Ζ Ζ Z eh Z EH Z Z z <
Ζ Q E Q Z Q Z Q Z Z
3 « < ω Eh fa < fa < Q
Ζ fa Z Pu Z fa Z fa z ω
Q Ζ Q Z Q Z Q Z Q fa
cd Ζ Pd Z 0 Z Pd Z Cd Z
ω ω ω en en fa 0 CQ 0 Z
Λ Ζ < h Z <3 i-* Z < Z Z < H 0
Ζ S Ζ h S ί EH g < z g z Z Z >
tou fa O U fa α o fa Oi 0 fa s <
Ρί μΐ X Λ u x z z x z z X cd οι ω
χ x x X X
Η < EH Z fa
Η fa PU Z H
Η ω o O 0 H Z 0
ω 0 Q z 0 z en 0 0 z
ω ζ Cd > z > 0 Z 0 >
Pd > S pu 3 Z z > Z 0
ζ 0 en M Z Q z en Z Q
hH Ο H < S < H Q fa z
ω < > X >1 en < < X
Ζ Z Oi Z z
ο 0 0 0 0
PU < fa < fa < fa < fa <
< > < > < > < > < >
Ο Ζ oi fa Ol fa Ol z Ol fa
Pd Μ Cd οι Pd fa Z fa cd fa
X Pd fa Pd fa Cd X z fa cd
Β οι 3 M 3 fa 3 O 3 fa
(d 0 0 Pd 0
Z S 2J fa
Æ Z l-H
fa X kj X
Η Q Z H 0
Ζ U Z U >-3 O Z U Z O
ο ω 0 ω 0 en 0 0 0 0
Ο Ζ 0 z 0 Z 0 Z 0 Z
e cd 0 0 Pd en 0 Cd Z 0 Cd en 0 Cd 0
0 ζ 0 m μι fa en Z fa en z ω 0 Z fa
ω ω ζ fa 0 z w en eh fa 0 H fa 0 0
> 0 Ζ > o z > Q Cd > 0 Z >00
Ζ 0 0 Z 0 0 Z 0 0 Z 0 0 Z 0 0
οι fa 0 O fa en oi < en O» fa 0 oi < 0
> οι < > Ot Eh > Ot < > Ol < > Oi <
ω > < fa > < ω > >i fa > fi ω > fi
Μ H H f—1 H
Η X. H X. Μ X H X, JH X
CQ οο CQ vr m en m σι 0 00
fa Ο fa o fa O fa O fa O
ü m ο 0 Q H 0 fa CM 0 0 0 0 fa
ω en cm fa en cm fa en CM fa en <m fa en cm
> CO Η > oo H > oo H > 00 H > C0 H
Table 3: Sequence IDs and AA sequences of monovalent receptor-blocking anti-VEGF VHHs (FR, framework; CDR, complementary determining région) SEQ ID NO: 9-46
Eu Eu EH Eu Eu
> > > > >
CM CM CM CM CM
H H Eu Eu Eu
CD 0 0 0 0
cm 0 O en CM 0 CM 0 CM 0
pi 0 0 0 ω 0 0 0 0 0 0
S & > 3 > 3 > B > B >
< 0
3 A A A
a Pi Pi Pi Pi
a XI A A A
ni Pi ci Pi pi
en en 0 0 0
en en 0 0 0
CD X 0 x 0 X 0 X < x
tn X Q X Q X Q X Q X Q
Pi FÉ X x rf X < X < X
Q Pu H Ci H a h Ci A a eu
U en Q en Q 0 Q 0 Q 0 Q
> < > < > < > < > <
Eu Eu Eh Eh Eh Eh Eu Eu H H
Z Q Z Ω z û Z Q Z Q
S ta a ω S M ta ω ta a
Eu CU eu eu Eu eu eu eu eu a
z ta Z ta Z E z ta z ta
Q XI Q XI Q A Q A Q A
ci en a 0 < a 0 rf Pi 0 rt Pi 0
en eu en eu < 0 Cu < 0 z < 0 eu <
H H CD H Eu o H H U t-H H U H Eu 0
ΟΊ H CM t* Eu Ο X Eu CM X Eu CM X Eu CM X
pi a A X ta xi x ia a x a xi x a a x
E Ci Q tu x > a x > Pi X > Pi x >
Hi X 1-H z x
X 0 H X 0 X 0 H
ο ω a 0 0 ω z ω 0 0
0 XI 0 a 0 A 0 A 0 CM
en en 0 XI 0 0 0 0 0 A
en > en en 0 > 0 > 0 0
K en en en > 0 0 0 0 0 >
Ω M Q H < H Q 1-H Q H 0
U < X < Q < X < X < Q
M ω ω ω a
a ta ta ta ta
0 0 0 0 0
CM CM o cm CM
< > < > < > < > < >
O > CM > CM > CM > CM >
fd a E ni E oi E pi a a a
K a ω a ω ia w a a a a
E B ci 3 E 3 Pi B Pi b a
rH S Ξ S S S
Pi en en 0 0 0
Ω X x X X X
U en 0 <n < 0 0 0 0 0 0
0 XI en 0 A en 0 a w 0 A 0 0 A 0
0 ta a 0 S a 0 S a 0 ni a 0 E 0
0 XI E-I 0 A Fl 0 A Eu 0 A EU 0 A Eu
en en ni en en Pi 0 0 Pi 0 0 Ci 0 0a
ω q 0 ω o 0 a q 0 a q 0 a Q 0
> 0 en > 0 en >00 >00 >00
A a a a a a xi eu < a a <
H o o < CM CM < CM CM < CM CM rtj CM CM >
a > > U > > u > > 0 > > 0 > > U
E a A 0 a a 0 W A 0 a a 0 a a 0
o rH CM 0
ô H <Λ H rH H rH H H 1-H H
Q Z H \ H X. HH \ H \ H \
H 9 m o PP rH PQ CO m r~- CQ M·
Ci4 rH £îq rH a O a o a 0
S G 0 < 0 < 0 m 0 0 0 a
UJ ω cm ω cm ω cm a cm a cm
> « > CM > CM > CM > CM > CM
H Eh F F F F F
> > > > > > >
Ο es Oi Oi es Oi es
Eh Eh Eh F F F F
0 0 0 0 0 0 0
ο ω es en OI CO o en OI CD Oi 0 OI 0
0 0 0 en 0 CO 0 en 0 CD 0 0 0 0
& > B > B > B > B > B > B >
Du <
►fa >fa ►fa i_5 ►fa ►fa
ai oi oi ci 2 Di Pi
►fa ►fa ►fa ►fa ►fa fal fal
2 Pi Pi Pi 2 Du Du
0 en en en en 0 0
0 en en en co 0 0
0 >4 0 0 >4 0 >4 0 X 0 >4 0 >4
>4 Q >H Q >4 Q >4 Q >4 [d >4 Q >4 Q
>h < >< λ; >h rf >H >4 >4
2 eh 2 Fl 2 F 2 F 2 F 2 F 2 F
0 Q CO Q en o en q CD Q 0 Cl 0 Q
> < > < > < > < > < > < > <
Eh F F H F F F F F F F F F F
Z Q Z Q z O Z Q Z o Z Q Z Q
z ω z ω Z [fa z ω S [d Z ω 2 Z
Eh 04 Eh 04 F Du en eu < eu F Pu F Pu
Z W Z Z Z Z z z z z Z Z Z Z
Q ni Q fal Q fal Q fal Q fal Q fal Q fal
fa en < Pi en < ci en «; Pi en < Z en co Pi 0 Æ fa en ρζ
ω pu < en fa < en z < CO fa PÇ 0 Z < en fa pi en fa pî
HH Eh U HH Eh O M 1—1 O fal F U fal 1—1 0 n F U H F O
EH CS >H F CS >4 F Ol >4 F Oi >4 F O< >4 F Oi >i F Oi >h
fa H [ï4 hfa >4 ω >fa >4 fa fal >4 tu fal >4 Z fal >4 tu fal >4
Pi >H > Pi >h > Ci >4 > Pi >4 > Pi >4 > Pi >4 > Pi >4 >
>4 >4 >4 HH >4 >4
H H Z >4 0 2 M H
>4 0 Z 0 >4 0 0 [fa >4 0 tu 0 >4 0
0 C» 0 ω 0 ω 0 fal 0 [d 0 W 0 es
0 fai 0 (fa 0 fal <n en 0 fal 0 fal 0 fal
0 en en en z en en > Z en 0 0 0 0
en > en > en > en en CD > 0 > 0 >
n en H < i-i en fai Q ►H en 1-1 < fal 0
< Q < Q < Q < >H < o < ο < Q
[£1 [fa [fa [fa [d [d td
Z z Z Z z ÎYn Z
0 0 0 0 0 0 0
CS es CS ot ot Q1 Qi
< > < > < > < > < > < > < >
Ot > oi > CS > OI > OI & o< > CS >
oi tu ci Eu pi Eu oi Eu Pi tu pi Eu Pi Eu
tu N Z Cd fai [J Pu [fa tu [d tu [d tu ω
B Pi B Ci B Pi B Pi B oi B Di B Du
2 S s s s S S
en en co co ω 0 0
>H >H >4 >4 >4 >4
en 0 en 0 en 0 en 0 en 0 0 < 0 0
0 J en 0 fal 0 O »fa en 0 fal 0 O fa en 0 fal 0 0 fal 0
0 Z tu 0 2 tu 0 2 tu 0 Z Z 0 Pi Z 0 2 tu 0 2 0
0 fa] H O fal Eh 0 fal F 0 fal F 0 fal F 0 ifa F Cd fal F
en en oi en ω ci en en z co en ci co en pi 0 0 Pi 0 0 ci
Pd Q 0 [d Q 0 ω q 0 td Q 0 [d Q 0 0 Q 0 W Q 0
> 0 en > 0 en > 0 en > 0 en > 0 en >00 >00
fa fa < fa fa fa fa F PC fa fa fa ►fa F > fa fa fa fal Z <
O CS > Oi OI < O Oî > CS CS < CS CS Z es es < CS OI >
> > U > > U > > 0 > > U > > U > > U > > U
U fa M [d fal 0 z fa) 0 z fal 0 td fal CD fa fa en ω fai 0
'T in CO e- CD ch 0
Ht 1-1 H 1-1 H i-l H H fal H H rd M CM
H \ H \ fal \ fal \ H \ fa4 \ fal \
PQ en CQ CD pp co PQ en CO 4V PQ 1—1 m 0
Z O Z O Z O Z O tu O tu t—1 tu 0
0 < 0 < O < 0 < 0 CQ 0 Q 0 Cd
ω en [fa en ω en Cd en M en [d 0 Cd 0
> CM > CM > CM > CM > CM > CM > CM
H EH EH Eh μ μ μ
> > > > > > >
Ο oi Ol Ol Ol
Ε-ι Eh EH Eh μ μ μ
0 0 0 0 0 ο 0
O en θ' en Oi en Oi en οι en o» en οι en
Ο en 0 en 0 en O en 0 en O en 0 en
Β > B > S > B > B > B > B >
0 < Cu < <
μ μ μ μ μ
ta E ta ta ta ta ta
μ μ μ μ μ μ μ
ta E ta ta ta ta ta
en en en en en 0 en
ω en en en en en en
Ο >1 0 x 0 x 0 x 0 x o x 0 x
X ο X Q X Q X Q x o X Q X Q
< X al x < x x < x < X < x
ta Η E Eh ta Eh ta μ ta μ ta μ ta μ
en Q en Q en Q en 0 en Q en o CO Q
> < > < > < > < > < > fi > <
ΕΗ ΕΗ EH EH Eh H Eh Eh H μ μ μ μ μ
J5 Ο Z Q Z ο Z Q Z Q Z Q Z Q
£ ω £ td ta ω ta ta ta ta οι M £ w
&Η Pt Eh ta EH ta en eu μ eu μ eu μ ta
ζ μ Z td Z td Z ta Z ta Z ta Z ta
Ο ta ο μ ο μ ο μ ο μ ο μ Q ta _
(Ü « rf oi en n; ta en fî ta en ti ta en < ta en fî ta eo ri
ω < en z < en ta <! en eu fi en eu < en z fi en eu fi
Η Εη Ο M H (J Ht μ O H H U μ μ u M H u μ μ O
μ Ο< X Eh ο X Eh Ol X μ θ' X μ Ol X μ O< X μ Ol X
fa μι χ ta μ x ta μ x ta μ x ta μ x ta μ x ta μ X
ta X Ο ta x > ta X Q ta x > ta X > ta x > ta x >
Η td H 1—1 X x x
X Ο X 0 X 0 X 0 H ta μ
Ο Μ Z ta 0 ta 0 ta X 0 X 0 X 0
ο μι 0 μ 0 μ 0 μ 0 Ol 0 ta 0 Ol
en en en en 0 en en en 0 μ 0 μ 0 μ
ω > en > en > en > en en en en en en
ω en en en en en en en en > en > en >
Ht Ο H Q H Q HH Û h en μ en h en
< X < X < x < X < Q < Q < Q
W ta w ta ω ta ta
td td ta ta ta ta Ol
0 0 0 0 0 0 0
οι oi Ol Ol Ol Ol Ol
< r* < > < > < > i > < > < r·
οι > oi > Ol > Ol > Ol > ot > Ol >
Ρί ta ta ta ta ta ta ta ta ta ta ta ta ta
ta ω ta ta ta H ta ta ta ta ta ta ta ta
Β ta B ta B ta B ta B ta b ta B ta
S s s Ξ Ξ S s
en en en en en en en
X X x x x x
en ο en o en 0 en 0 en 0 en 0 en 0
ο μ ω 0 μ en 0 μ en ο μ ο 0 μ en Ο μ en 0 μ en
0 ta ta 0 ta ta 0 ta ta 0 ta ta 0 ta en o ta ta ο ta en
ο μ εη 0 μ eh ο μ h 0 μ eh 0 μ μ 0 μ μ ο μ μ
en en ta en en ta en en ta en en ta en en ta eo en ta en en ta
ω ο 0 ta Q 0 ta Q 0 ta Q 0 ta Q 0 ta Q 0 ta Q 0
> 0 en > 0 en > O en > 0 en > 0 en > O en > 0 en
μ eu ta h-1 rt< rtj μ ta ta μ eu ta μ ta fi μ μ < μ eu <
οι οι < O O < θ' θ' fiC ο o < Ol Ol > οι ot E Ol o» >
S > ο > > U > > O > > O > > U > > ο > > u
ω μ en w μ en ω μ en ω μ en ω μ en ω μ en ω μ en
γ4 CM en in ν> x
H CM H CM H CM H CM Ht CM μ CM Ht CM
Η X H X H X Ht X Ht X μ x Ht X
CQ CM en m en μ ta en ta n* m œ ω x
ta ο ta o ta x ta o ta O ta o ta O
0 ta 0 ta 0 ta 0 0 0 U 0 Q 0 0
ta m ω en ta en ta en ta ’O’ ta X ta x
> CM > CM > CM > CM > CM > CM > CM
H Εη εη Εη Η 0
> > > > > > >
Ο 0 οι οι ΟΙ Oi Oi
H Εη ΕΗ ΕΗ ΕΗ H 0
0 0 0 0 0 0 0
ο ω ΟΙ 0 ΟΙ 0 ΟΙ 0 ΟΙ 0 Ol 0 Oi 0
ο ω 0 0 0 0 0 0 0 0 0 0 O 0
3 > 3 > 3 > 3 > 3 > 3 > 3 >
0 0
μι 0 0 0 0 0
0 0 0 0 0 0 0
0 μι 0 0 0 0 0
0 0 0 0 0 0 0
0 0 ω 0 0 0 0
ω 0 ω 0 0 0 0
0 X 0 X 0 X 0 X 0 X 0 X 0 X
X Ο X Ο X Q X Ο X Q X Q X Q
rf! X < X < X λ; χ rf X x < X
0 Η 0 ΕΗ 0 Εη 0 ÊH 0 Εη 0 H 0 0
0 Ο 0 Ο 0 Q 0 Ο 0 Ο 0 Q 0 Q
> < > < > < > < > < > < > <
Η Ε-ί Εη Η ΕΗ ΕΗ Εη 0 Εη Εη Eh H < 0
Ζ Ο Z Ο ζ ο Ζ ο ζ ο Z O Z Q
ίχ ω 2 ω 2 0 2 0 2 0 X 0 0 0
Ε-ι Ο ΕΗ 0 Εη 0 Ε-ι Cm Εη 0 Eh 0 0 0
ζ ιχ Z ÎX Ζ 2 Ζ X Ζ Ζ Z 2 z tx
Ο 0 ο μι Ο 0 Ο 0 Ο 0 Q 0 Q 0
0 0 < £Χ 0 < 0 0 < 0 0 < 0 0 < 0 0 < 0 0 <
co eu < ω ζ < 0 0 iÇ 0 0 < 0 ζ fi 0 0 fi 0 0 fi
Μ EH υ Μ Η 0 Η Εη 0 Η Εη 0 Η Η 0 H 0 U n 0 U
Η Ο X Η Ο< X 0 ΟΙ X Εη ΟΙ >Η Η Ο X H ΟΙ X 0 Ol x
0 μ X 2 0 X 0 0 X 0 0 X 0 0 X 0 0 X 0 0 X
0 X > 0 X > 0 X Q 0 X Ο 0 X > 0 X > 0X0
X 2 Η X ίΧ X M
Η X 0 X 0 X 0 >ι 0 X X 0
X 0 Ζ 0 0 0 0 0 Ζ 0 0 0 0 0
0 ο 0 μ 0 0 0 0 0 0 0 0 0 0
υ μ 0 0 0 0 0 0 0 0 0 0 0 0
ω co 0 > ω > ω > 0 < 0 0 0 >
0 > 0 0 ω ω 0 0 0 0 0 > 0 0
Η 0 Η1 Q n Ο H Q Μ Ο H < X Q
< Ο < X < X < X < X < Q < X
ω 0 0 0 0 0 0
ο IX 2 0 ζ 0 IX
0 0 0 0 ο 0 0
ο οι οι ΟΙ οι Ol oi
< > < > < > < > < > < > < >
ΟΙ > οι > οι > ΟΙ > ΟΙ > oi > ot >
0 0 0 0 0 0 0 0 0 0 0 0 0 0
ιχ ω 0 0 0 0 0 Μ 0 0 0 0 0 0
3 (X 3 0 3 0 3 0 3 0 3 0 3 0
3 3 3 3 3 3 S
co 0 0 0 0 0 0
X X X X X X X
ω 0 0 0 0 0 0 0 0 0 0 < 0 0
0 μ ω 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 O 0 0
ο 2 0 Ο 0 0 0 0 0 0 X X 0 0 0 0 0 0 0 !X 0
Ο 0 Η 0 0 ΕΗ 0 0 Εη 0 0 ÊH 0 0 Η 0 0 Eh 0 0H
0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0
Μ Ο 0 0 Q 0 0 Ο 0 0 Q 0 0 Q 0 0 Q 0 0 Q 0
>00 >00 >00 >00 >00 >00 >00
μι ίχ < 0 < < 0 Εη rtj 0 0 0 0 < fi 0 0 0 1-4 CM Γχ-ι
ΟΙ ΟΙ Η Ο» ΟΙ rt! ΟΙ ΟΙ ΰΐ Ο ΟΜ 0 οι fi oi ot < OI Ol <
> > ο > > Ο > > ο > > Ο > > ο > > U > > U
ω μι 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0
00 en ο γΗ CM en
Μ CM l-Η CM Η 0 η en χ en h en h en
Μ \ Ι-Η \ Μ X. Η X. H \ H \ H \
0 σι m ο- 0 Γ- 0 CM 0 0 CM ffl C-
ιχ ο 0 Ο 0 Ο 0 Ο 0 o 0 O 0 O
0 Ο 0 0 0 0 0 0 0 0 0 0 0 0
0 Ο 0 ο 0 <Η 0 σ 0 σ 0 O 0 O
> 0 > 0 > 0 > 0 > 0 > >
A Eh EH EH A A A
> > > > > > >
Ot Ot Ot Ot ot Ot Ot
É“* Eh Eh A A A A
0 O 0 0 0 0 0
O en ot en Ot 0 Ot 0 ot 0 Ot 0 Ot 0
0 en 0 en 0 0 0 0 0 0 0 0 0 0
B > B > B > B > B > B > B >
g rf PC
ni ni Cu ne A A
A Al A A A A A
ni ni en 2 A A A
en en 0 0 0 0 0
en en 0 0 0 0 0
0 x 0 X 0 X 0 X 0 X 0 X 0 X
X 0 x o X Q X Q X Q X Q X Q
pC X PC X PC X PC X pC X PC X PC X
A Eh Ci Eh A eh 2 A A A A A A A
en 0 en Q en o 0 Q 0 Q 0 O 0 Q
> < > pC > pC > pC > < > pC > <
EH EH Eh A A Eh A A Eh Eh A A a fi
Z 0 Z 0 z 0 Z Q Z Q Z Q Z Q
S 0 Z M E 0 A ω A 0 A 0 A 0
En PU H CU Eh CU A cu A A A A A A
Z A z te Z A S A Z A Z A Z A
Q A 0 Al 0 A Q A Q A Q A Q A
ni en pC A en < ni 0 pC A 0 pC A 0 PC A 0 pC A 0 pC
en a fi 0 a fi 0 A < 0 eu pC 0 A pC 0 A fi 0 A pC
H En o HEU MAO M S U MAU MAO MAO
A Ot X EH ot X H ΟΙ X A Ot X A ΟΙ X A Ot X A Ot X
0 A X lu ι-l X tu A X A A X A A X 0 A X 0 A X
A X 0 A x > A x > A X 0 A X > A X > A X Q
H X x H H X H
X O H H X 0 X 0 I—ί X 0
ο 0 tu 0 tu 0 0 ω 0 0 X 0 0 0
o a 0 0 0 ta 0 A 0 A 0 Ot 0 A
en en 0 Al 0 A 0 0 0 0 0 A 0 0
en > en en 0 0 0 > 0 > 0 0 0 >
en en en > 0 > 0 0 0 0 0 > 0 0
M 0 M < H pC H Q H Q M 0 M Q
pC X < 0 pC Q pC X pC X pC Q < X
0 0 0 ω 0 0 0
A A A A A A A
0 0 0 0 0 0 0
ot Ot ot ot Ot Ot Ot
PC > pC > PC > PC > pC > < £ PC >
ot > o > ot > O > ot > Ot > OI >
ni Eu ne L· ni a A Eu A 0 A Eu A 0
t 0 tu 0 tu 0 a ω 0 0 0 0 0 0
B ni S ni B ni B A B A B A B A
3 3 3 3 3 > 3
en 0 0 0 0 0 0
X x x X X X X
en 0 en 0 0 pC 0 0 0 0 0 O 0 0
0 A 0 0 A 0 0 A 0 0 A 0 0 A 0 0 A 0 0 A 0
0 S tu 0 A tu ο A A 0 A tu 0 A 0 0 A 0 0 A A
0 q h 0 A Eh 0 A A 0 A A 0 A A 0 A A 0 A A
en en ni 0 0 a 0 0 ne 0 0 A 0 0 A 0 0 A 0 0 A
0 0 0 000 ω o 0 0 Q 0 0 Q 0 0 Q 0 0 0 0
3 0 en >00 >00 >00 >00 >00 >00
Al eu tu A eu tu A eu tu A A A A A 0 A A pC A A 0
ot ot < ot ot PC ot Ot pC Ot Ο pC Ot Ot pC O O» > Ot Ot pC
> > u > > U > > u > > U > > U > > U > > U
ω ai en ω a 0 M A 0 0 A 0 0 A 0 0 A 0 0 A 0
m co X CD en O rd
ai en H (VJ H 0 H 0 M 0 m n· M
Al \ Η ·Χ H X. X M \ M \ Μ X
en o CQ m 0 0 CQ 0 0 0 CQ rH 0 M
tu <—I tu o tu O 0 O 0 O Ph rH 0 M
0 X 0 0 0 0 0 pC 0 Q 0 0 0 0
0 o 0 <-t 0 t—i 0 (XI 0 CN 0 CN 0 <O
> 'T > 'PT > sr > -PT > sr > > 0
H Eh Eh Eh Eh
> > > > >
Ο ex ex ex ex
H Eh Eh Eh Eh
0 0 0 0 0
ex CO ex en ex en ex en ex en
o co 0 en 0 en 0 en 0 en
B > B > B > B > B >
< < < 0
fa fa fa fa fa
Cd ed od Od 2
fa fa fa fa fa
ai Bd od 2 2
en en ω en en
en en en en en
0 >-1 < >4 0 >4 0 >4 0 >4
>4 Q >H Q >4 Q >4 Q >4 Q
< >h < >4 < >4 f£ >4 2 >4
2 H 2 EH Cd ÉH Cd £h 2 Eh
en Q en Q en Q en Q en Q
> < > < > < > < > <
Eh Eh Eh H En Eh En Eh EH EH
Z Q Z Q S û Z Q Q Q
cd ω fa fa fa fa fa fa fa fa
Eh CU Eh BU Eh 0-i EH DU Eh eu
Z fa Z fa Z fa Z fa z fa
Q fa Q fa Q fa Q fa Q fa
ai en rf cd en < cd en Æ od en Æ cd en <
en eu 2 en eu < en eu 2 en z 2 en fa <
h h υ hH EH O fa Eh u H H o H Eh 0
Eh O >H Eh CX >H Eh Ο X Eh ο >4 En CX >4
h μ) fa fa Sh fa fa >4 fa fa >4 fa fa >H
(U >4 Û ai >h > cd >4 > cd >4 > ed >4 >
H >H fa fa >4
>H 0 fa > 0 >4 0 fa
0 [£] >4 0 0 fa Z fa >4 0
0 1-1 0 fa 0 fa 0 fa 0 fa
en en 0 fa en en en en 0 fa
en > en en en > en > en en
en en en > en en en en < >
H Q H < H Q FH O fa <
< >4 < Q < >4 < >4 < Q
fa fa fa H
Ξ fa fa fa fa
0 0 0 0 0
ex ex ex ex ex
< > < > < > < > < >
ex t» ex > ex > ex > ex >
dû Eu Od fa Dd fa cd fa ai fa
fa ω fa fa fa fa fa w fa w
B Cd B Od B cd B od B od
Ξ S s S S
en en en en en
>4 >4 >4 >H >4
en 0 en 0 en 0 en 0 en 0
0 fa en 0 fa en Q fa en 0 fa en 0 fa en
0 S fa 0 od fa 0 fa fa 0 cd fa 0 fa fa
0 U Eh 0 fa EH 0 fa Eh fa fa Eh 0 fa eh
en co ed en en od en ω ed en en od en en cd
fa Q 0 M 0 0 fa Q 0 fa Q 0 H Q 0
> 0 en > 0 en > 0 en > 0 en > 0 en
fa eu fa fa < fa fa eu < H-| rtj 1—| Uq Llq
ex ex < ex ex < CX ex < ex ex 2 ex ex <
> > 0 > > U > > 0 > > 0 > > 0
ω fa en ω fa en ω fa en fa fa en M fa en
CM en m CD
H H fa 'r fa fa M1
H «s. H \ H \ H \ fa \
CQ 03 tn ï—i ea σ> en o OQ CM
fa O fa O fa o fa fa fa O
0 < 0 < 0 < 0 Q 0 fa
fa O ω fa fa CM fa CM W CM
> CD > CD > cd > eo > CD
Dissociation rates of inhibitory VHHs are analyzed on Biacore (Biacore T100 instrument, GE Healthcare). HBS-EP+ buffer is used as running buffer and experiments are performed at 25°C. Recombinant human VEGF 165 is irreversibly captured on a CM5 sensor chip via amine coupling (using EDC and NHS) up to a target level of +/-1500RU. After immobilization, surfaces are deactivated with 10 min injection of 1M ethanolamine pH8.5. A référencé surface is activated and deactivated with respectively EDC/NHS and ethanolamine. Periplasmic extracts of VHHs are injected at a 10-fold dilution in running buffer for 2 min at 45pl/min and allowed to dissociate for 10 or 15 min. Between different samples, the surfaces are regenerated io with régénération buffer. Data are double referenced by subtraction of the curves on the référencé channel and of a blank running buffer injection. The of the processed curves is evaluated by fitting a two phase decay model in the Biacore T100 Evaluation software v2.0.1. Values for kj-fast, kj-slow and % fast are listed in Table
4.
Table 4: Off-rate détermination of anti-VEGF receptor-blocking VHHs with Biacore
B-cell lineag e Unique sequence variant Représentative VHH ID kd(fast) kd(slow) % fast Binding level (RU)
1 1 VEGFBI122B07 1.50E-02 7.80E-05 31 328
1 2 VEGFBH23A08 1.30E-02 5.00E-05 19 502
1 3 VEGFBII23B04 8.80E-03 4.00E-05 12 768
1 4 VEGFBII27D08 2.40E-02 8.10E-05 13 225
1 5 VEGFBII24C04 1.30E-02 3.40E-05 17 456
1 6 VEGFBII27G07 1.30E-02 3.80E-05 18 471
1 7 VEGFBII22E04 1.80E-02 1.10E-04 14 520
1 8 VEGFBII23A03 1.50E-02 3.20E-05 15 487
1 9 VEGFBII22B06 3.80E-02 9.00E-05 23 168
1 10 VEGFBII23A09 2.70E-02 4.60E-05 20 247
1 11 VEGFBII23G03 2.80E-02 8.60E-05 28 141
1 12 VEGFBII22A11 2.20E-02 4.70E-05 12 461
1 13 VEGFBII23A06 1.70E-02 3.70E-05 13 547
1 14 VEGFBII23F11 2.70E-02 1.30E-04 22 134
1 15 VEGFBII22A10 3.70E-02 4.00E-05 19 229
1 16 VEGFBII23F05 1.60E-02 1.30E-04 29 198
1 17 VEGFBII23D11 1.90E-02 5.80E-05 13 510
1 18 VEGFBII23F02 n/d n/d n/d n/d
1 19 VEGFBII23E05 1.50E-02 6.90E-05 18 275
1 20 VEGFBII31C07 3.70E-02 1.50E-04 25 77
1 21 VEGFBII30C09 1.50E-02 7.60E-05 19 264
1 22 VEGFBII30E07 1.70E-02 1.30E-04 29 226
1 23 VEGFBII39G04 1.40E-02 7.40E-04 40 210
1 24 VEGFBII41G03 1.20E-02 2.70E-04 20 332
1 25 VEGFBII41B05 1.90E-02 1.20E-04 16 324
1 26 VEGFBII40F02 1.20E-02 9.80E-05 20 258
1 27 VEGFBII39E02 1.90E-02 2.40E-04 13 181
1 28 VEGFBII42D05 3.30E-02 1.50E-04 26 77
1 29 VEGFBII40G07 1.80E-02 3.20E-04 19 139
1 30 VEGFBII42A05 1.60E-02 3.40E-04 25 118
1 31 VEGFBI142F11 9.10E-03 5.00E-04 46 100
1 32 VEGFBII40H10 1.40E-02 2.90E-04 17 200
1 33 VEGFBII62A09 4.10E-02 1.10E-04 23 84
1 34 VEGFBII60A09 3.70E-02 9.30E-05 20 106
1 35 VEGFBÜ62F02 1.40E-02 8.50E-05 21 205
1 36 VEGFBII62D10 1.90E-02 1.60E-04 40 94
1 37 VEGFBII61A01 7.40E-03 1.70E-04 21 275
1 38 VEGFBII56E11 3.30E-02 1.40E-04 24 76
n/d, not determined
Example 5
Characterization of purified anti-VEGF VHHs
Three inhibitory anti-VEGF VHHs are selected for further characterization as purified s protein: VEGFBII23B04, VEGFBII24C4 and VEGFBII23A6. These VHHs are expressed in E. coli TG1 as c-myc, His6-tagged proteins. Expression is induced by addition of 1 mM IPTG and allowed to continue for 4 hours at 37°C. After spinning the cell cultures, periplasmic extracts are prepared by freeze-thawlng the pellets. These extracts are used as starting material for VHH purification via IMAC and size io exclusion chromatography (SEC). Final VHH préparations show 95% purity as assessed via SDS-PAGE.
5.1 Evaluation of human VEGF165/VEGFR2 blocking VHHs in human VEGF165/human VEGFR2-Fc blocking ELISA
The blocking capacity of the VHHs is evaluated in a human VEGF165/human VEGFR2-Fc blocking ELISA. In brief, 1 pg/mL of VEGFR2-Fc chimera (R&D Systems, Minneapolis, MN, USA) is coated in a 96-well MaxiSorp plate (Nunc, Wiesbaden, Germany). Dilution sériés (concentration range 1 mM - 64pM) of the purified VHHs in PBS buffer containing 0.1% casein and 0.05% Tween 20 (Sigma) are incubated in the presence of 4 nM biotinlyated VEGF165. Residual binding of bio-VEGF165 to VEGFR2 is detected using horseradish peroxidase (HRP) conjugated extravidin (Sigma, St Louis, MO, USA) and TMB as substrate. As contrais Bevacizumab (Avastin®) and Ranibizumab (Lucentis®) are taken along. Dose inhibition curves are shown in Figure 1 ; the corresponding IC50 values and % inhibition are summarized in Table 5.
Table 5: IC50 (nM) values and % inhibition for monovalent VHHs in hVEGF165/hVEGFR2-Fc compétition ELISA
VHH ID IC50 (nM) % inhibition
VEGFBII23B04 2.1 100
VEGFBII23A06 3.0 100
VEGFBII24C04 2.5 100
Ranibizumab 1.6 100
Bevacizumab 1.7 100
5.2 Evaluation of human VEGF165/VEGFR2 blocking VHHs in human VEGF165/human VEGFR1-Fc blocking ELISA
VHHs are also evaluated in a human VEGF165/human VEGFR1-Fc blocking ELISA. In brief, 2 pg/mL of VEGFR1-Fc chimera (R&D Systems, Minneapolis, MN, USA) is coated in a 96-well MaxiSorp plate (Nunc, Wiesbaden, Germany). Dilution sériés (concentration range 1 mM - 64pM) of the purified VHHs in PBS buffer containing 0.1% casein and 0.05% Tween 20 (Sigma) are incubated in the presence of 0.5nM biotinlyated VEGF165. Residual binding of bio-VEGF165 to VEGFR1 is detected using horseradish peroxidase (HRP) conjugated extravidin (Sigma, St Louis, MO,
USA) and TMB as substrate. As contrais Bevacizumab, Ranibizumab and an irrelevant VHH (2E6) are taken along. Dose inhibition curves are shown in Figure 2; the corresponding IC50 values and % inhibition are summarized in Table 6.
Table 6: IC50 (nM) values and % inhibition of monovalent VHHs in hVEGF165/hVEGFR1-Fc compétition ELISA
VHH ID ICSo(nlVI) % inhibition
VEGFBII23B04 0.5 64
VEGFBII23A06 0.9 55
VEGFBII24C04 0.8 71
Ranibizumab 1.2 91
Bevacizumab 1.5 96
5.3 Evaluation ofthe anti-VEGF165 VHHs in the human VEGF165/human VEGFR2Fc blocking AlphaScreen
The blocking capacity of the VHHs is also evaluated in a human VEGF165/human VEGFR2-Fc blocking AlphaScreen. Briefly, serial dilutions of purified VHHs (concentration range: 200 nM - 0.7 pM) in PBS buffer containing 0.03 % Tween 20 (Sigma) are added to 4pM bio-VEGF165 and incubated for 15 min. Subsequently VEGFR2-Fc (0.4 nM) and anti-Fc VHH-coated acceptor beads (20 pg/ml) are added and this mixture is incubated for 1 hour in the dark. Finally, streptavidin donor beads (20 pg/ml) are added and after 1 hour of incubation in the dark, fluorescence is measured on the Envision microplate reader. Dose-response curves are shown in the Figure 3. The IC50 values for VHHs blocking the human VEGF165 - human VEGFR2-Fc interaction are summarized in Table 7.
Table 7: IC50 (pM) values and % inhibition for VHHs in hVEGF165/hVEGFR2-Fc compétition AlphaScreen
VHH ID IC50 (pM) % inhibition
VEGFBII23B04 160 100
VEGFBII23A06 250 100
VEGFBII24C04 250 100
Ranibizumab 860 100
5.4 Evaluation ofthe anti-VEGF165 VHHs in the human VEGF165/human VEGFR 1Fc blocking AlphaScreen
The blocking capacity of the VHHs is also evaluated in a human VEGF165/human s VEGFRI-Fc blocking AlphaScreen. Briefly, serial dilutions of purified VHHs (concentration range: 500 nM - 1.8 pM) ) in PBS buffer containing 0.03 % Tween 20 (Sigma) are added to 0.4 nM bio-VEGF165 and incubated for 15 min. Subsequently VEGFR1-Fc (1 nM) and anti-Fc VHH-coated acceptor beads (20 pg/ml) are added and this mixture is incubated for 1 hour in the dark. Finally, streptavidin donor beads io (20 pg/ml) are added and after 1 hour of incubation in the dark, fluorescence is measured on the Envision microplate reader. Dose-response curves are shown in the Figure 4. The IC50 values and % inhibition for VHHs blocking the human VEGF165 human VEGFR1-Fc interaction are summarized in Table 8.
Table 8: IC50 (nM) values for VHHs in hVEGF165/hVEGFR1-Fc compétition
AlphaScreen
VHH ID IC50(nM) % inhibition
VEGFBII23B04 0.9 41
VEGFBII23A06 0.4 46
VEGFBII24C04 0.2 53
Ranibizumab 3.3 79
5.5 Détermination ofthe affinity ofthe human VEGF165-VHH interaction
Binding kinetics of VHH VEGFBII23B04 with hVEGF165 is analyzed by SPR on a
Biacore T100 instrument. Recombinant human VEGF165 is immobilized directly on a
CM5 chip via amine coupling (using EDC and NHS). VHHs are analyzed at different concentrations between 10 and 360nM. Samples are injected for 2 min and allowed to dissociate up to 20 min at a flow rate of 45 μΙ/min. In between sample injections, the chip surface is regenerated with 100 mM HCl. HBS-EP+ (Hepes buffer pH7.4 + s EDTA) is used as running buffer. Binding curves are fitted using a Two State
Reaction model by Biacore T100 Evaluation Software v2.0.1. The calculated affinities of the anti-VEGF VHHs are listed in Table 9.
Table 9: Affinity KD (nM) of purified VHHs for recombinant human VEGF165
VEGF165
VHH ID ka ka1 ka2 (Mis1) kd (s’1) kdi (s'1) kd2 (s1) KD (nM)
VEGFBII23B04(a) - 2.1E+05 1.4E-02 - 8.6E- 03 2.4E-04 0.7
VEGFBII23A06(a) - 4.2E+05 2.0E-02 - 5.7E- 02 1.0E-04 0.7
VEGFBII24C04(a> - 3.2E+05 1.8E-02 - 2.6E- 02 9.6E-05 0.4
(a) Heterogeneous binding curve resulting in no 1:1 fit, curves are fitted using a Two State Reaction model by Biacore T100 Evaluation Software v2.0.1
5.6 Binding to mouse VEGF164
Cross-reactivity to mouse VEGF164 is determined using a binding ELISA. In brief, recombinant mouse VEGF164 (R&D Systems, Minneapolis, MS, USA) is coated ovemight at 4°C at 1 pg/mL in a 96-well MaxiSorp plate (Nunc, Wiesbaden, Germany). Wells are blocked with a casein solution (1 % in PBS). VHHs are applied as dilution sériés (concentration range: 500nM - 32pM) in PBS buffer containing 0.1% casein and 0.05% Tween 20 (Sigma) and binding is detected using a mouse anti-myc (Roche) and an anti-mouse-HRP conjugale (DAKO) and a subséquent enzymatic reaction in the presence of the substrate TMB (3,3',5,5'tetramentylbenzidine) (Pierce, Rockford, IL, USA) (Figure 5). A mouse VEGF164 reactive mAb is included as positive control. As référencé, binding to human VEGF165 is also measured. EC50 values are summarized in Table 10.
Table 10: EC50 (pM) values for VHHs in a recombinant human VEGF165 and mouse VEGF164 binding ELISA
rhVEGF165 rmVEGF164
VHH ID EC50 (pM) ECS0 (pM)
VEGFBII23B04 297 NB
VEGFBII24C04 453 NB
VEGFBII23A06 531 NB
NB, no binding
5.7Binding to VEGF121
Binding to recombinant human VEGF121 is assessed via a solid phase binding ELISA. Briefly, recombinant human VEGF121 (R&D Systems, Minneapolis, MS, USA) is coated overnight at 4°C at 1 pg/mL in a 96-well MaxiSorp plate (Nunc, Wiesbaden, Germany). Wells are blocked with a casein solution (1% in PBS). VHHs are applied as dilution sériés (concentration range: 500nM - 32pM) in PBS buffer containing 0.1% casein and 0.05% Tween 20 (Sigma) and binding is detected using a mouse anti-myc (Roche) and an anti-mouse-HRP conjugate (DAKO) and a subséquent enzymatic reaction in the presence of the substrate TMB (3,3’,5,5’tetramentylbenzidine) (Pierce, Rockford, IL, USA) (Figure 6). As positive control serial dilutions of the VEGFR2 is taken along. EC5o values are summarized in Table 11.
Table 11: EC50 (pM) values for monovalent VHHs in a recombinant human VEGF121 binding ELISA
VHH ID ECso (pM)
VEGFBII23B04 510
VEGFBII24C04 792
VEGFBII23A06 928
5.8 Binding to VEGF family members VEGFB, VEGFC, VEGFD and PIGF
Binding to VEGFB, VEGFC, VEGFD and PIGF is assessed via a solid phase binding ELISA. In brief, VEGFB, VEGFC, VEGFD and PIGF (R&D Systems, Minneapolis,
MS, USA) are coated overnight at 4°C at 1 pg/mL in a 96-well MaxiSorp plate (Nunc, 15 Wiesbaden, Germany). Wells are blocked with a casein solution (1% in PBS). VHHs are applied as dilution sériés (concentration range: 500nM - 32pM) and binding is detected using a mouse anti-myc (Roche) and an anti-mouse-AP conjugate (Sigma, St Louis, MO, USA). As positive contrais serial dilutions of the appropriate receptors are taken along and detected with horseradish peroxidase (HRP)-conjugated goat 20 anti-human IgG, Fc spécifie antibody (Jackson Immuno Research Laboratories Inc.,
West Grave, PA, USA) and a subséquent enzymatic reaction in the presence of the substrate TMB (3,3’,5,5'-tetramentylbenzidine) (Pierce, Rockford, IL, USA). Doseresponse curves of VHHs and contrais are shown in Figure 7. The results show that there was no détectable binding of the selected VHHs to VEGFB, VEGFC, VEGFD or 25 PIGF.
5.9 Epitope binning
Biacore-based epitope binning experiments are performed to investigate which VEGF binders bind to a similar or overlapping epitope as VEGFBII23B04. To this end, VEGFBII23B04 is immobilized on a CM5 sensorchip. For each sample, human VEGF165 is passed over the chip surface and reversibly captured by VEGFBII23B4. Purified VHHs (100 nM) or periplasmic extracts (1/10 diluted) are then injected with a surface contact time of 240 seconds and a flow rate of 10 uL/minute. Between different samples, the surface is regenerated with régénération buffer (100 mM HCl). Processed curves are evaluated with Biacore T100 Evaluation software. VHHs could be divided within two groups: group one which gave additional binding to VEGFBII23B04 captured VEGF165 and a second group which is not able to simultaneously bind to VEGFBII23B04 captured VEGF165. Table 12-A summarizes the binding epitopes of the tested VHHs.
The same assay set-up is used to assess whether VEGFR1, VEGFR2, Ranibizumab and Bevacizumab are able to bind to human VEGF-165 simultaneously with VEGFBII23B04. Table 12-B présents the additional binding responses to VEGFBII23B04-captured VEGF165. Only VEGFR2 is not able to bind to VEGFBII23B04-captured VEGF165, underscoring the blocking capacity of VEGFBII23B04 for the VEGF-VEGFR2 interaction. In addition, these data show that the VEGFBII23B04 epitope is different from the Bevacizumab and Ranibizumab epitope.
Table 12-A: Epitope binning of anti-VEGF VHHs - simultaneous binding with VEGFBII23B04
No or low 1C02 1E07 4B08 8E07 8F07 12A07 12B01 86C11 86F11 86G08
additional 86G10 86G11 87B07 88A01 88A02 88B02 88E02 88G03 88G05 88G11
binding to 23B04-
captured VEGF165* 88H01 89B04 89D04 89F09 89G09 89H08 24C04 23A6 27G07 23B04
Additional 3D12 5B02 5B03 5B05 6G02 7D08 8D09 8F06 10C07 10E07
binding to 23B04-
captured VEGF165 10G04 10G05 11C08 11D09 11E04 11E05 11F12 86H09 41C05
* indicating same or overlapping epitopes
Table 12-B: Epitope binning of VEGFBII23B04- binding of benchmark inhibitors or cognate receptors on VEGFBII23B04 captured VEGF165
Injection step Binding [sample] Binding level (RU)
1 VEGF165 100 nM 1727
2 VEGFBII23B04 100 nM -
3 Ranibizumab 100 nM 763
4 Bevacizumab 100 nM 1349
5 VEGFR1 100 nM 1011
6 VEGFR2 100 nM -
5.10 Characterization of the anti-VEGF VHHs in the HUVEC prolifération assay io The potency of the selected VHHs is evaluated in a prolifération assay. In brief, primary HUVEC cells (Technoclone) are supplement-starved over night and then 4000 cells/well are seeded in quadruplicate in 96-well tissue culture plates. Cells are stimulated in the absence or presence of VHHs with 33ng/mL VEGF. The prolifération rates are measured by [3H] Thymidine incorporation on day 4. The results of the HUVEC prolifération assay are shown in Table.
Table 13: IC50 (nM) values and % inhibition of monovalent VEGFBII23B04,
VEGFBII23A06 and VEGFBII24C04 in VEGF HUVEC prolifération assay
VHH ID ICso(nM) % inhibition
VEGFBII23B04 0.36 91
Bevacizumab 0.21 92
VHH ID ICSO (nM) % inhibition
VEGFBII23A06 4.29 73
VEGFBII24C04 3.8 79
Bevacizumab 0.78 78
5.11 Characterization ofthe anti-VEGF VHHs in the HUVEC Erk phosphorylation assay
The potency ofthe selected VHHs is assessed in the HUVEC Erk phosphorylation assay. In brief, primary HUVE cells are serum-starved over night and then stimulated io in the absence or presence of VHHs with 10ng/mL VEGF for 5 min. Cells are fixed with 4% Formaldéhyde in PBS and ERK phosphorylation levels are measured by ELISA using phosphoERK-specific antibodies (anti-phosphoMAP Kinase pERK1&2, M8159, Sigma) and polyclonal Rabbit Anti-Mouse-Immunoglobulin-HRP conjugate (PO161, Dako). As shown in Table 14, VEGFBII23B04 and Bevacizumab inhibit the is VEGF induced Erk phosphoryaltion by at least 90%, with IC50s <1 nM.
Table 14: IC50 (nM) values and % inhibition of monovalent VEGFBII23B04 in VEGF
HUVEC Erk phosphorylation assay
VHH ID ICso(nM) % inhibition
VEGFBII23B04 0.37 90
Bevacizumab 0.63 98
Example 6
Génération of multivalent anti-VEGF blocking VHHs
VHH VEGFBII23B04 is genetically fused to either VEGFBII23B04 resulting in a homodimeric VHH (AA sequence see Table 15) or different VEGF binding VHHs resulting in heterodimeric VHHs. To generate the heterodimeric VHHs, a panel of
10 unique VEGF binding VHHs are linked via a 9 or 40 Gly-Ser flexible linker in two different orientations to VEGFBI123B04 (AA sequences see Table 15). Homodimeric VEGFBII23B04 (VEGFBII010) and the 40 heterodimeric bivalent' VHHs are expressed in E. coli TG1 as c-myc, His6-tagged proteins. Expression is induced by addition of 1 mM IPTG and allowed to continue for 4 hours at 37°C. After spinning the cell cultures, periplasmic extracts are prepared by freeze-thawing the pellets. These extracts are used as starting material and VHHs are purified via IMAC and desalting resulting in 90% purity as assessed via SDS-PAGE.
100
Table 15: Sequence ID, VHH ID and AA sequence of bivalent anti-VEGF VHHs (each of the used linkers is highlighted in one relevant sequence)
Q X
X >
O o m
LL
CD
LU >
ω CD O) 'ίο CD
101
Ο) 4· ο CÛ
σι I ^Γ Ο CÛ
Ο)
I ο CÛ
102
EVQLVESGGGLVQTGDSLRLSCEVSGRTFSSYSMGWFRQAQGKEREFWAISKGGYKYDSVSLEG rftiskdnakntvylqinslkpedtavyycassraygssrlrladtyeywgqgtqvtvssggggsg| gggsggggsggggsggggsggggsggggsggggsevqlvesggglvqpggslrlscaasgsa vgditvawyrqapgiqrqlvatitpsgytyywdfvkgrftisrdnsknivylqmnslkpedtaayycn TQFYWGQGTQVTVSS evqlvesggglvqtgdslrlscevsgrtfssysmgwfrqaqgkerefwaiskggykydsvsleg rftiskdnakntvylqinslkpedtavyycassraygssrlrladtyeywgqgtqvtvssggggsg gggsggggsggggsggggsggggsggggsggggsevqlvesggglaqagdslrlscaasgrs fshynmgwfrqapgkerefvasirggggsttyansvkdrftisrenakntvylqmnslkpedtavy ycaatafyrgpydydywgqgtqvtvss EVQLVESGGGLVQTGDSLRLSCEVSGRTFSSYSMGWFRQAQGKEREFWAISKGGYKYDSVSLEG RFTISKDNAKNTVYLQINSLKPEDTAVYYCASSRAYGSSRLRLADTYEYWGQGTQVTVSSGGGGSG GGGSGGGGSGGGGSGGGGSGGGGSGGGGSGGGGSEVQLVESGGGLVQPGGSLRLSCVASGIR FMSMAWYRQAPGKHRELVARISSGGTTAYVDSVKGRFTISRDNSKNTVYLQMNSLKAEDTAVYYCN TFSSRPN PWGAGTQVTVSS EVQLVESGGGLVQTGDSLRLSCEVSGRTFSSYSMGWFRQAQGKEREFWAISKGGYKYDSVSLEG RFTISKDNAKNTVYLQINSLKPEDTAVYYCASSRAYGSSRLRLADTYEYWGQGTQVTVSSGGGGSG GGGSGGGGSGGGGSGGGGSGGGGSGGGGSGGGGSEVQLVESGGGLVQPGGSLRLSCAASGNI FSNNAMAWYRQAPGKQRELVARISSGGGFTYYLDSVKGRFTVSRDNAKNTVYLQMNSLKPEDTAVY YCNAAYRTYNYWGQGTQVTVSS EVQLVESGGGLVQTGDSLRLSCEVSGRTFSSYSMGWFRQAQGKEREFWAISKGGYKYDSVSLEG RFTISKDNAKNTVYLQINSLKPEDTAVYYCASSRAYGSSRLRLADTYEYWGQGTQVTVSSGGGGSG GGGSGGGGSGGGGSGGGGSGGGGSGGGGSGGGGSEVQLVESGGGLVQAGGSLRLSCAASGRT FSNYAMGWFRQAPGKERVLVADISSSGINTYVADAVKGRFTiSRDNAKNTVYLQMNSLKPEDTAVYY CAASAWWYSQMARDNYRYWGQGTQVTVSS
VEGFBII021 VEGFBÜ023
VEGFBII23B04- 40GS-4B08/139 VEGFBII23B04- 40GS-5B03/140 VEGFBH23B04- 40GS-5B05/141 VEGFBII23B04- 40GS-6G02/142 VEGFBII23B04- 40GS-10E07/143
103
LU >
m
104
105
106
EVQLVESGGGLVQPGGSLRLSCAASGNIFSNNAMAWYRQAPGKQRELVARISSGGGFTYYLDSVKG RFTVSRDNAKNTVYLQMNSLKPEDTAVYYCNAAYRTYNYWGQGTQVTVSSGGGGSGGGGSGGGG SGGGGSGGGGSGGGGSGGGGSGGGGSEVQLVESGGGLVQTGDSLRLSCEVSGRTFSSYSMGW FRQAQGKEREFWAISKGGYKYDSVSLEGRFTISKDNAKNTVYLQINSLKPEDTAVYYCASSRAYGSS RLRLADTYEYWGQGTQVTVSS EVQLVESGGGLVQAGGSLRLSCAASGRTFSNYAMGWFRQAPGKERVLVADISSSGINTYVADAVKG RFTISRDNAKNTVYLQMNSLKPEDTAVYYCAASAWWYSQMARDNYRYWGQGTQVTVSSGGGGSG GGGSGGGGSGGGGSGGGGSGGGGSGGGGSGGGGSEVQLVESGGGLVQTGDSLRLSCEVSGRT FSSYSMGWFRQAQGKEREFWAISKGGYKYDSVSLEGRFTISKDNAKNTVYLQINSLKPEDTAVYYC ASSRAYGSSRLRLADTYEYWGQGTQVTVSS ÈVQLVESGGGLVQPGGSLRLACAASGFTLSSSWMYWVRQAPGKGLEWVSRISPGGLFTYYVDSVK GRFSVSTDNANNTLYLQMNSLKPEDTALYSCAKGGAPNYTPRGRGTQVTVSSGGGGSGGGGSGG GGSGGGGSGGGGSGGGGSGGGGSGGGGSEVQLVESGGGLVQTGDSLRLSCEVSGRTFSSYSM GWFRQAQGKEREFWAISKGGYKYDSVSLEGRFTISKDNAKNTVYLQINSLKPEDTAVYYCASSRAY GSSRLRLADTYEYWGQGTQVTVSS EVQLVESGGGLVQAGDSLRLSCTASGRTFNSYAMGWFRQAPGKERESVAHINRSGSSTYYADSVK GRFTISRDNAKNTVYLQLNSLKPEDTAVYYCAAGRYYSSDGVPSASFNYWGQGTQVTVSSGGGGS GGGGSGGGGSGGGGSGGGGSGGGGSGGGGSGGGGSEVQLVESGGGLVQTGDSLRLSCEVSGR TFSSYSMGWFRQAQGKEREFWAISKGGYKYDSVSLEGRFTISKDNAKNTVYLQINSLKPEDTAVYY CASSRAYGSSRLRLADTYEYWGQGTQVTVSS I EVQLVESGGGLVQAGGSLRLSCTASGSAFKSYRMGWFRRTPGKEDEFVASISWTYGSTFYADSVK GRFTMSRDKAKNAGYLQMNSLKPEDTALYYCAAGAQSDRYNIRSYDYWGQGTQVTVSSGGGGSG GGGSGGGGSGGGGSGGGGSGGGGSGGGGSGGGGSEVQLVESGGGLVQTGDSLRLSCEVSGRT FSSYSMGWFRQAQGKEREFWAISKGGYKYDSVSLEGRFTISKDNAKNTVYLQINSLKPEDTAVYYC ASSRAYGSSRLRLADTYEYWGQGTQVTVSS
VEGFBII025
VEGFBI16G02-40GS- 23B04/162 VEGFB1I10E07- 40GS-23B04/163 VEGFB1I12B01- 40GS-23B04/164 VEGFBII86C11- 40GS-23B04/165 VEGFBII86H09- 40GS-23B04/166
107
EVQLVESGGGLVQPGGSLKLSCTASGFTFSTSWMHWVRQAPGKGLEWVSSIPPVGHFANYAPSVK GRFTISRDNAKNTLFLQMNSLKSEDTAVYYCAKDSAGRTKGQGTQVTVSSGGGGSGGGGSGGGGS GGGGSGGGGSGGGGSGGGGSGGGGSEVQLVESGGGLVQTGDSLRLSCEVSGRTFSSYSMGWF RQAQGKEREFWAISKGGYKYDSVSLEGRFTISKDNAKNTVYLQINSLKPEDTAVYYCASSRAYGSS RLRLADTYEYWGQGTQVTVSS EVQLVESGGGLVQAGGSLRLSCAASERTFSNYAMDWFRQAPGKEREFVAAITRSGGGTYYADSVK GRFTISRDNAKNTVYLQMNSLKPEDTAVYYCAATRSSTIWGVGGMEYWGKGTQVTVSSGGGGSG GGGSGGGGSGGGGSGGGGSGGGGSGGGGSGGGGSEVQLVESGGGLVQTGDSLRLSCEVSGRT FSSYSMGWFRQAQGKEREFWAISKGGYKYDSVSLEGRFT1SKDNAKNTVYLQINSLKPEDTAVYYC ASSRAYGSSRLRLADTYEYWGQGTQVTVSS
VEGFBII87B07- 40GS-23B04/167 VEGFBI188A01- 40GS-23B04/168
108
The panel of 40 bivalent VHHs is tested in the VEGFR2 and VEGFR1 blocking AlphaScreen assay, as described in Example 5.3 and 5.4, respectively. Based on potency and maximum level of inhibition, the 5 best bivalent VHHs (VEGFBII021, VEGFBII022, VEGFBI023, VEGFBI024 and VEGFBII025) are chosen for further characterization. An overview of the screening results for the 5 selected bivalent VHHs in the compétitive VEGFR2 and VEGFR1 AlphaScreen is shown in Table 16.
Table 16: Potency and efficacy of 5 best bivalent VHHs in the VEGF/VEGFR1 and VEGF/VEGFR2 compétition AlphaScreen assay
VHH ID VEGFR2 VEGFR1
IC50 (pM) IC50 (pM) % inhibition
VEGFBII021 9 16 100
VEGFBII022 7 8 100
VEGFBII023 38 44 91
VEGFBII024 12 46 100
VEGFBII025 51 39 82
Example 7
Characterization of formatted anti-VEGF VHHs
VHHs VEGFBII010, VEGFBII021, VEGFBII022, VEGFBII023, VEGFBII024 and VEGFBII025 are compared side-by-side in the VEGFR2 and VEGFR1 blocking
ELISA (Figure 8 and 9, Table 17 and Table 18 respectively) and AlphaScreen assay (Figure 10 and 11, Table 19 and 20) as described in Examples 5.1, 5.2, 5.3 and 5.4, respectively.
109
Table 17: IC50 (pM) values and % inhibition for formatted VHHs in hVEGF165/hVEGFR2-Fc compétition ELISA
VHH ID IC50 (pM) % inhibition
VEGFBII010 49 100
VEGFBII021 204 100
VEGFBII022 164 100
VEGFBII023 213 100
VEGFBÎI024 292 100
VEGFBII025 577 100
Bevacizumab 315 100
Ranibizumab 349 100
Table 18: IC50 (pM) values and % inhibition of formatted VHHs in
VEGF165/hVEGFR1-Fc compétition ELISA
VHH ID IC50 (PM) % inhibition
VEGFBII010 73.5 67
VEGFBII021 254 97
VEGFBII022 225 89
VEGFBII023 279 91
VEGFBII024 326 92
VEGFBII025 735 91
Bevacizumab 484 91
Ranibizumab 594 96
110
Table 19: IC50 (pM) values and % inhibition for formatted VHHs in hVEGF165/hVEGFR2-Fc compétition AlphaScreen
VHH ID IC50 (PM) % inhibition
VEGFBII010 16 100
VEGFBII021 7 100
VEGFBII022 7 100
VEGFBII023 46 100
VEGFBII024 50 100
VEGFBII025 51 100
Ranibizumab 600 100
Table 20: IC50 (pM) values and % inhibition of formatted VHHs in VEGF165/hVEGFR1-Fc compétition AlphaScreen
VHH ID IC50 (pM) % inhibition
VEGFBII010 21 70
VEGFBII021 12 100
VEGFBII022 9 98
VEGFBII023 48 87
VEGFBII024 69 98
VEGFBII025 71 82
Ranibizumab 1300 87
In addition, formatted VHHs are also tested for their capacity to block the mVEGF164/mVEGFR2-huFc interaction. In brief, serial dilutions of purified VHHs (concentration range: 4μΜ - 14.5 pM) in PBS buffer containing 0.03 % Tween 20
111 (Sigma) are added to 0.1 nM biotinylated mVEGF164 and incubated for 15 min. Subsequently mouse VEGFR2-huFc (0.1 nM) and anti-huFc VHH-coated acceptor beads (20 pg/ml) are added and this mixture is incubated for 1 hour. Finally, streptavidin donor beads (20 pg/ml) are added and after 1 hour of incubation fluorescence is measured on the Envision microplate reader. Dose-response curves are shown in Figure 12. The IC50 values for VHHs blocking the mouse VEGF164/VEGFR2-hFC interaction are summarized in Table 21.
Table 21: IC50 (pM) values and % inhibition forformatted VHHs in mVEGF164/mVEGFR2-hFc compétition AlphaScreen
VHH ID IC50 (nM) % inhibition
VEGFBII022 108 100
VEGFBII024 - -
mVEGF164 0.05 100
Ranibizumab - -
The formatted VHHs are also tested in ELISA for their ability to bind mVEGFI 64 and human VEGF165 (Example 5.6; Figure 13; Table 22); VEGF121 (Example 5.7; Figure 15; Table 23) and the VEGF family members VEGFB, VEGFC, VEGFD and
PIGF (Example 5.8; Figure 14). Binding kinetics for human VEGF165 are analyzed as described in Example 5.5. The Kd values are listed in Table 24.
Table 22 EC50 (pM) values forformatted VHHs in a recombinant human VEGF165 and mouse VEGF164 binding ELISA
rhVEGF165 rmVEGF164
VHH ID EC50 (pM) ECS0 (pM)
VEGFBII010 428 -
VEGFBII021 334 502
VEGFBII022 224 464
VEGFBII023 221 -
VEGFBII024 320 -
VEGFBII025 668 -
112
Table 23: EC5o (pM) values for formatted VHHs in a recombinant human VEGF121 binding ELISA ___________________________________
rhVEGF121
VHH ID EC5o (pM)
VEGFBII010 920
VEGFBII022 540
VEGFBII024 325
VEGFBII025 475
Table 24: Affinity KD (nM) of purified formatted VHHs for recombinant human VEGF165
VHH ID ka1 (1/Ms) kdi (1/s) ka2 (1/S) kd2(1/s) KD (nM)(a)
VEGFBII010 4.5E+05 1.7E-02 2.9E-02 1.3E-04 0.16
VEGFBII021tb) 1.2E+06 1.1E-02 2.3E-02 1.9E-04 0.07
VEGFBII022lb) 1.2E+06 9.1E-03 1.4E-02 2.6E-04 0.14
VEGFBII023(b) 3.0E+05 1.8E-02 2.4E-02 2.7E-04 0.69
VEGFBII024lb) 3.0E+05 1.3E-02 2.6E-02 2.8E-04 0.47
VEGFBII025lb) 3.3E+05 1.7E-02 1.8E-02 3.7E-04 1.1
<a,KD=kd1/ka1*(kd2/(kd2+ka2)) (W Curves are fitted using a Two State Reaction model by Biacore T100 Evaluation Software v2.0.1
VHHs VEGFBII010, VEGFBII022, VEGFBII024 and VEGFBII025 are also tested in the VEGF-mediated HUVEC prolifération and Erk phosphorylation assay.
The potency ofthe selected formatted VHHs is evaluated in a prolifération assay. In brief, primary HUVEC cells (Technoclone) are supplement-starved over night and is then 4000 cells/well are seeded in quadruplicate in 96-well tissue culture plates. Cells are stimulated in the absence or presence of VHHs with 33ng/mL VEGF. The prolifération rates are measured by [3H] Thymidine incorporation on day 4. The
113 results shown in Table 25 demonstrate that the formatted VHHs and Bevacizumab inhibitthe VEGF-induced HUVEC prolifération by more than 90%, with IC50S <1nM.
Table 25: IC50 (nM) values and % inhibition of formatted VHHs in VEGF HUVEC prolifération assay
VHH ID ICSo(nM) % inhibition
VEGFBII010 0.22 95
VEGFBII021 0.40 98
VEGFBII022 0.34 100
VEGFBII023 0.52 98
VEGFBII024 0.38 96
VEGFBII025 0.41 104
Bevacizumab 0.21 92
The potency of the selected formatted VHHs is assessed in the HUVEC Erk phosphorylation assay. In brief, primary H U VE cells are serum-starved over night and 10 then stimulated in the absence or presence of VHHs with 10ng/mL VEGF for 5 min.
Cells are fixed with 4% Formaldéhyde in PBS and ERK phosphorylation levels are measured by ELISA using phosphoERK-specific antibodies (anti-phosphoMAP Kinase pERK1&2, M8159, Sigma) and polyclonal Rabbit Anti-MouseImmunoglobulin-HRP conjugate (PO161, Dako). As shown in Table 26, the formatted
VHHs and Bevacizumab inhibit the VEGF induced Erk phosphoryaltion by more than 90%, with IC50S <1nM.
114
Table 26: IC50 (nM) values and % inhibition of formatted VHHs in VEGF HUVEC Erk phosphorylation assay
VHH ID IC50(nM) % inhibition
VEGFBII010 0.19 92
VEGFBII021 0.21 103
VEGFBII022 0.18 94
VEGFBII023 0.25 100
VEGFBII024 0.23 94
VEGFBII025 0.23 99
Bevacizumab 0.63 98
Example 8
Sequence optimization
8.1 Sequence optimization of VEGFBII23B04
The amino acid sequence of VEGFBII23B04 is aligned to the human germline sequence VH3-23/JH5, see Figure 16 (SEQ ID NO: 179)
The alignment shows that VEGFB1I23B04 contains 19 framework mutations relative to the référencé germline sequence. Non-human residues at positions 14,16, 23, 24, 41, 71,82, 83 and 108 are selected for substitution with their human germline counterparts. A set of 8 VEGFBII23B04 variants is generated carrying different combinations of human residues at these positions (AA sequences are listed in Table 27). One additional variant is constructed in which the potential isomerization site at position D59S60 (CDR2 région, see Figure 16, indicated as bold Italie residues) is removed by introduction of a S60A mutation.
115
Table 27: AA sequence of sequence-optimized variants of VHH VEGFBII23B04 (FR, framework; CDR, complementary determining région)
116
WGQGTLVTV SS WGQGTLVTV SS
æ £ g 5 CD j— cd h g 3
< ai > < ai x
(Z -i LU £Z _l LU
ω tz x 0 tz >
z Z
O σ
> w > W
C < > <
P ° P °
z t: z
A ζ A C
Z £ 9 Q a S! § i 9 Q UJ $2 Q.
lr * H (Z
U- -1 P —1
(Z 0 tZ 0
A , , A , ,
X W X U-|
8 S % ω § g £ ω
S2 o,„ - Q
< X 0
Lk LL
LU LU
tZ tz
LU LU
A A
0 0
CL < CL <
g σ [Z
U- ·>
Z %
S 5
0 0
X „
0 c 0 0
0 _j 0 0 _l 0
0 £Z p 0 £Z U-
O _! h- 0 -J h
OT 0 ai w 0 tr
LU Q 0 UJ 0 0
>00 >00
-J CL >
O O LU ο a w
> > 0 > > 0
LJ J W LL J W
(N CO
EGFBII 14C09/5 _ «o Cû § LL 2 0 9 tu ?
> T- > x-
WGQGTLVTV SS WGQGTQVTV SS
0 |_ 0 5 0 < CD μ CD h 0 y
X _3 > □
< ai > < a >
£Z -1 LU Cl —l LU
ω a: x 0 (Z x
z z
σ σ
> w X w
s < > <
P ° P 0
z £ Z £
a ς A C
ί 1 A ° « s g É « £
tz 5 t s
ai ω ai 0
A A , .
X LU X w
§g A 0 § g A i
0 Q 0 Q
< > 0 < > 0
LL LL
LU UJ
ai 01
LU LU
A k:
0 0
a < a «
<3 tz a tz
5 5 δ δ
Ξ Ξ
0 0
X „
0 0 0 0
0 _j 0 0 _J 0
0 tz p 0 ai p
0 —i H 0 -j H
(0 0 ÛC 0 0 tz
LU O 0 UJ O 0
>00 >00
ο σ < o a lu
> > o > > 0
IL J W LL J «
xr U)
_ «n — !9
m 2 CD o
LL 2 0 g LU ? EGF 18E1
> V > <“
117
These variants are characterized as purified proteins in the VEGF165/VEGFR2 AlphaScreen (Example 5.3, Figure 17). The melting température (Tm) of each clone is determined in a thermal shift assay, which is based on the increase in fluorescence signal upon incorporation of Sypro Orange (Invitrogen) (Ericsson et al, Anal. Biochem. 357 (2006), pp289-298). Ail variants displayed comparable IC50 when compared to VEGFBII23B04 and Tm values which are similar or higher when compared to the parental VEGFBII23B04. Table 28 summarizes the IC50 values and Tm values at pH 7 for the 9 clones tested.
Table 28: IC50 (pM) values, % inhibition and melting température (@pH 7) of sequence-optimized variants of VEGFBII23B04
VHHID ICso(pM) % inhibition Tm ® pH 7 (°C)
VEGFBII23B04 (wt) 169 100 63
VEGFBII111D05 209 100 68
VEGFBII111G06 366 100 71
VEGFBII112D11 221 100 70
VEGFBII113A08 253 100 69
VEGFBII113E03 290 100 68
VEGFBII114C09 215 100 71
VEGFBII114D02 199 100 74
VEGFBII114D03 227 100 64
VEGFBII118E10 189 100 62
In a second cycle, tolerated mutations from the humanization effort (VEGFBII111G06) and mutations to avoid potential posttranslational modification at selected sites (the D16G, the S60A substitution and an E1D mutation) are combined resulting in a sequence-optimized clone derived from VEGFBII23B04: VEGFBII0037. One extra sequence-optimized variant (VEGFBII038) is anticipated which contains the same substitutions as VEGFBII0037, with the exception of the I82M mutation, as this mutation may be associated with a minor drop in potency. The sequences from both sequence-optimized clones are listed in Table 29. VEGFBII0037 and VEGFBII0038 are characterized in the VEGF165/VEGFR2 blocking AlphaScreen (Example 5.3, Figure 18), the melting température is determined in the thermal shift assay as described above and the affinity for
118 binding on VEGF165 is determined in Biacore (Example 5.5). An overview of the characteristics of the 2 sequence-optimized VHHs is presented in Table 30.
Table 29: AA sequences of sequence-optimized variants of VHH VEGFBII23B04
VHH ID/ SEQ 1D NO: FR1 CDR 1 FR2 CDR 2 FR3 CDR 3 FR 4
VEGFBII0 37 56 DVQLVES GGGLVQ PGGSLRL SCAASG RTFS SYS MG WFRQA PGKER EFVV AISKG GYKYD AVSLE G RFTISRDNAKN TVYLQMNSLRP EDTAVYYCAS SRAYGSS RLRLADTY EY WGQG TLVTV SS
VEGFBIIO 38 57 DVQLVES GGGLVQ PGGSLRL SCAASG RTFS SYS MG WFRQA PGKER EFVV AISKG GYKYD AVSLE G RFTISRDNAKN TVYLQINSLRP EDTAVYYCAS SRAYGSS RLRLADTY EY WGQG TLVTV SS
Table 30: IC50 (pM) values, % inhibition, melting température (@pH 7) and affinity (pM) of sequence-optimized clones VEGFBII037 and VEGFBII038
VHH ID IC50 (PM) % inhibition Tm (°C) @ pH 7 KD (pM)
VEGFBH23B04 152 100 63 560
VEGFBII037 300 100 72 270
VEGFBII038 143 100 71 360
8.2 Sequence optimization of VEGFBII5B05
The amino acid sequence of VEGFBI15B05 is aligned to the human germline sequence VH3-23/JH5, see Figure 19 (SEQ ID:NO: 179 The alignment shows that VEGFBII5B05 contains 15 framework mutations relative to the référencé germline sequence. Non-human residues at positions 23, 60, 83,105, 108 are selected for
119 substitution with their human germline counterparts while the histidine at position is selected for substitution by glutamine. One humanization variant is constructed carrying the 6 described mutations (AA sequence is listed in Table 31).
Table 31: AA sequences of sequence-optimized variants of VHH VEGFBII5B05 (FR, framework; CDR, complementary determining région)
VHH ID/ SEQ ID NO: FR1 CD R1 FR2 CDR2 FR3 CDR3 FR4
VEGFBII119 G11/ 125 EVQLVES GGGLVQ PGGSLR LSCAAS GIRFM SM A WYRQAP GKQREL VA RISS GGTT AYAD SVKG RFTISRDNS KNTVYLQM NSLRAEDTA VYYCNT FSSRP NP WGQG TLVTV SS
VEGFBII120 E10/ 126 EVQLVES GGGLVQ PGGSLR LSCVAS GIRFI SM A WYRQAP GKHREL VA RISS GGTT AYVD SVKG RFTISRDNS KNTVYLQM NSLKAEDTA VYYCNT FSSRP NP WGAG TQVTV SS
One additional variant is constructed in which the potential oxidation site at position M30 (CDR1 région, see Figure 19 indicated as bold italic residue) is removed by introduction of a M30I mutation. Both variants are tested for their ability to bind hVEGF165 using the ProteOn. In brief, a GLC ProteOn Sensor chip is coated with human VEGF165. Periplasmic extracts of the variants are diluted 1/10 and injected across the chip coated with human VEGF165. Off-rates are calculated and compared to the off-rates of the parental VEGFBII5B05. Off-rates from the 2 variants are in the same range as the off-rates from the parental VEGFBII5B05 indicating that ail mutations are tolerated (Table 32).
Table 32: Off-rates sequence-optimized variants VEGFBII5B05
VHH ID binding level (RU) kd(1/s)
VEGFBII5B05 242 6.15E-02
VEGFBII119G11 234 7.75E-02
VEGFBII120E10 257 4.68E-02
120
In a second cycle, mutations from the humanization effort and the M30I substitution are combined resulting in a sequence-optimized clone of VEGFBII5B05, designated VEGFBII032. The sequence is listed in Table 33. Affinity of VEGFBII032 is determined by Biacore (see Example 5.5) and the melting température is determined in the thermal shift assay as described above. An overview of the characteristics of the sequence-optimized VHH VEGFBII032 is presented in Table 34.
Table 33: AA sequence of sequence-optimized clone VEGFBII032 (FR, framework; CDR, complementary determining région)
VHH ID/ SEQID NO: FR1 CD R1 FR2 CDR2 FR3 CDR3 FR4
VEGFBIIO 32/ 127 EVQLVES GGGLVQ PGGSLR LSCAAS GIRFI SM A WYRQAP GKQREL VA RISS GGTT AYAD SVKG RFTISRDNS KNTVYLQM NSLRAEDTA VYYCNT FSSR PNP WGQGT LVTVSS
Table 34: Melting température (@pH 7) and affinity (nM) of sequence-optimized clone VEGFBII032
VHH ID Tm (°C) @ pH 7 KD (nM)
VEGFBII5B05(wt) 69 32
VEGFBII0032 71 44
The potency of the sequence-optimized clones VEGFBII037 and VEGFBII038 is evaluated in a prolifération assay. In brief, primary HUVEC cells (Technoclone) are supplement-starved over night and then 4000 cells/well are seeded in quadruplicate in 96-well tissue culture plates. Cells are stimulated in the absence or presence of VHHs with 33ng/ml_ VEGF. The prolifération rates are measured
121 by [3H] Thymidine incorporation on day 4. The results shown in Table 35, demonstrate that the activity (potency and degree of inhibition) of the parental VHH VEGFBII23B04 is conserved in the sequence optimized clone VEGFBII038.
Table 35: IC50 (nM) values and % inhibition of the sequence optimized clones VEGFBII037 and VEGFBII038 in VEGF HUVEC prolifération assay
VHH ID IC5o(nM) % inhibition
VEGFBII23B04 0.68 92
VEGFBII037 1.54 78
VEGFBII038 0.60 92
Bevacizumab 0.29 94
Example 9
Construction, production and characterization of bivalent VHHs targeting Ang2
VHHs 1D01 (SEQ ID No:214), 11B07, 00908 and 00027 (SEQ ID No:216) are genetically fused to 1D01 (SEQ ID No: 214), 11B07, 00908 and 00027 (SEQ ID No:216), respectively, resulting in homodimeric VHHs. The bivalent VHHs are linked via a 9-GlySer or 40-GlySer flexible linker. The encoding DNA sequences of the formatted VHHs are cloned in the expression vector pAX172. VHHs are expressed in Pichia pastoris as c-terminally myc - His6 tagged proteins. In brief, BGCM cultures are started from a single colony streak incubated over weekend at 30°C (250 rpm). After medium switch to BMCM, cultures are incubated until evening at 30°C (250 rpm) and followed by an induction with 100% methanol. The next day the cultures are induced an additional 3 times (morning, afternoon, evening). The next day cultures are centrifuged for 20 min at 4°C (1,500xg). The His6-tagged VHHs présent in the supernatant are purified through immobilized métal affinity chromatography (IMAC) followed by desalting (DS) and finally gel filtration (GF) to remove any endotoxins/impurities. An overview of the format and sequence of ail bivalent VHHs is depicted in Figure 20 and Table 36-A (linker
122 sequences underlined), SEQ ID Nos 180-185. Expression levels are indicated in Table 36-B.
To explore the anti-Ang2 blocking properties in comparison with the monovalent building blocks, bivalent VHHs are analyzed in a human Ang2/hTie2 (Figure 211), mouse Ang2/mTie2 (Figure 21-2), cyno Ang2/cTie2 (Figure 21-3) and human Ang1/hTie2 (Figure 22) compétition ELISA. A summary of IC50 values is shown in Table 37.
Table 36-A
Sequences of bivalent VHH targeting Ang2
VHH ID AA sequence
ANGBII00001 EVQLVE SGGGLVQAGGSLRLSCAAS G FT FDDYALGWFRQAAGKEREGVSCIRCS DGSTYYADSVKGR FTISSDNAKNTVYLQMNSLKPEDTAVYYCAASIVPRSKLEPYEYDAWGQGTLVTVSSGGGGSGGGSE VQLVESGGGLVQAGGSLRLSCAASGFTFDDYALGWFRQAAGKEREGVSCIRCSDGSTYYADSVKGRF TISSDNAKNTVYLQMNSLKPEDTAVYYCAASIVPRSKLEPYEYDAWGQGTLVTVSS (SEQ ID NO; 180)
ANGBII00002 EVQLVESGGGLVQAGGSLRLSCAASGFTFDDYALGWFRQAAGKEREGVSCIRCSDGSTYYADSVKGR FTISSDNAKNTVYLQMNSLKPEDTAVYYCAASIVPRSKLEPYEYDAWGQGTLVTVSSGGGGSGGGGS GGGGSGGGGSGGGGSGGGGSGGGGSGGGGSEVQLVESGGGLVQAGGSLRLSCAASGFTFDDYALGWF
RQAAGKEREGVSCIRCSDGSTYYADSVKGRFTISSDNAKNTVYLQMNSLKPEDTAVYYCAASIVPRS KLEPYEYDAWGQGTLVTVSS (SEQ ID NO: 181)
ANGBII00003 EVQLVESGGGLVQVGDSLRLSCAASGRTFSTYLMVGWFRQAPGKEREFAAGIWSSGDTAYADSVRGR FTISRDNAKNTVYLQMNS LKTE DTAVYYCAGSYDGNYYIPG FYKDWGQGTLVTVSSGGGGSGGGSEV QLVE SGGG LVQVGDSLRLSCAAS GRT FSTYLMVGWFRQAPGKERE FAAGIWS SGDTAYADSVRGR FT ISRDNAKNTVYLQMNSLKTEDTAVYYCAGSYDGNYYIPGFYKDWGQGTLVTVSS (SEQ ID NO: 182)
ANGBII00004 EVQLVE SGGGLVQAGGSLRLSCAASG FTLDDYAIGW FRQAPGKEREGVSSIRDNDGSTYYADSVKGR FTISSDNDKNTVYLQMNSLKPEDTAVYYCAAVPAGRLRFGEQWYPLYEYDAWGQGTLVTVSSGGGGS GGGSEVQLVESGGGLVQAGGSLRLSCAASGFTLDDYAIGWFRQAPGKEREGVSSIRDNDGSTYYADS VKGRFTISSDNDKNTVYLQMNSLKPEDTAVYYCAAVPAGRLRFGEQWYPLYEYDAWGQGTLVTVSS (SEQ ID NO: 183)
ANGBII00005 EVQLVESGGGLVQAGGSLRLSCAASGFTLDDYAIGWFRQAPGKEREGVSSIRDNDGSTYYADSVKGR FTISSDNDKNTVYLQMNSLKPEDTAVYYCAAVPAGRLRFGEQWYPLYEYDAWGQGTLVTVSSGGGGS GGGGS GGGGSGGGGSGGGGSGGGG S GGGGSGGGGSEVQLVE SGGGLVQAGGS LRLS CAASG FTLD DY
AIGW FRQAPGKEREGVSSIRDN DGSTYYADSVKGRFTISSDNDKNTVYLQMNSLKPE DTAVYYCAAV PAGRLRFGEQWYPLYEYDAWGQGTLVTVSS (SEQ ID NO: 184)
ANGBII00006 EVQLVESGGGLVQPGGSLRLSCAASGITLDDYAIGWFRQAPGKEREGVSSIRDNGGSTYYADSVKGR FTISSDNSKNTVYLQMNSLRPEDTAVYYCAAVPAGRLRYGEQWYPIYEYDAWGQGTLVTVSSGGGGS GGGSEVQLLESGGGLVQPGGSLRLSCAASGITLDDYAIGWFRQAPGKEREGVSSIRDNGGSTYYADS VKGRFTISSDNSKNTVYLQMNSLRPEDTAVYYCAAVPAGRLRYGEQWYPIYEYDAWGQGTLVTVSS (SEQ ID NO: 185)
123
Table 37: IC5o values (pM) in human Ang2/human Tie2, mouse Ang2/mouse Tie2, cyno Ang2/cyno Tie2 and hAng1/hTie2 compétition ELISA.
hAng2 mAng2 cAng2 hAng1/hAng2 ratio
VHH ID Format IC50 (pM) IC50 (PM) IC50 (pM)
1D01 6,973 10,455 9,484 >10,800
ANGBII00001 11 18 27 n.d.
ANGBII00002 20 32 n.d. n.d.
11B07 À 15,205 10,000 16,400 > 8,570
ANGBII00003 24 47 35 n.d.
00027 541 1,785 568 >14,878
ANGBII00004 17 33 34 n.d.
ANGBII00005 ^wa?7j|^ÿ^Îâo27j 13 32 n.d. n.d.
00908 52 85 79 >192,014
ANGBII00006 19 28 25 6,052,631
AMG386 4 3 10 5,600
124
n.d., not determined
Example 10
Construction, production and characterization of trivalent bispecific VHHs targeting VEGF and Ang2 using anti-serum abumin as half-life extension The anti-VEGF VHH VEGFBII00038 (US 2011/0172398 A1) and the anti-Ang2 VHH 00027 (SEQ ID No:216) are used as building blocks to generate bispecific VHHs VEGFANGBII00001-00004. A genetic fusion to a sérum albumin binding VHH is used as half-life extension methodology. Building blocks are linked via a triple Ala or 9 Gly-Ser flexible linker. VHHs are produced and purified as described in Example 9. An overview of the format and sequence of ail four bispecific VHHs is depicted in Figure 23 and Table 37-A (linker sequences underlined), SEQ ID Nos 186-189. Expression levels are indicated in Table 38-B.
Table 38-A
Sequences of bispecific VHH targeting VEGF and Ang2
VHH ID AA sequence
VEGFANGBII00001 DVQLVESGGGLVQPGGSLRLSCAASGRTFSSYSMGWFRQAPGKEREFWAISKGGYKYDAVSLEGRF TISRDNAKNTVYLQINSLRPEDTAVYYCASSRAYGSSRLRLADTYEYWGQGTLVTVSSGGGGSGGGS EVQLVE SGGGLVQPGNS LRLS CAASGFT FSS FGMSWVRQAPGKGLEWVS SISGSGS DT LYADSVKGR FTISRDNAKTTLYLQMNSLRPEDTAVYYCTIGGSLSRSSQGTLVTVSSGGGGSGGGSEVQLVESGGG LVQAGGSLRLSCAASGFTLDDYAIGWFRQAPGKEREGVSSIRDNDGSTYYADSVKGRFTISSDNDKN TVYLQMNSLKPEDTAVYYCAAVPAGRLRFGEQWYPLYEYDAWGQGTLVTVSS (SEQ ID NO: 186)
VEGFANGBII00002 EVQLVESGGGLVQAGGSLRLSCAASGFTLDDYAIGWFRQAPGKEREGVSSIRDNDGSTYYADSVKGR FTISSDNDKNTVYLQMNSLKPEDTAVYYCAAVPAGRLRFGEQWYPLYEYDAWGQGTLVTVSSGGGGS GGGSEVQLVESGGGLVQPGNSLRLSCAASGFTFSSFGMSWVRQAPGKGLEWVSSISGSGSDTLYADS VKGRFTISRDNAKTTLYLQMNSLRPEDTAVYYCTIGGSLSRSSQGTLVTVSSGGGGSGGGSDVQLVE SGGGLVQPGGSLRLSCAASGRTFSSYSMGWFRQAPGKEREFWAISKGGYKYDAVSLEGRFTISRDN AKNTVYLQINSLRPEDTAVYYCASSRAYGSSRLRLADTYEYWGQGTLVTVSS (SEQ ID NO: 187)
VEGFANGBII00003 EVQLVESGGGLVQAGGSLRLSCAASGFTLDDYAIGWFRQAPGKEREGVSSIRDNDGSTYYADSVKGR FTISSDNDKNTVYLQMNSLKPEDTAVYYCAAVPAGRLRFGEQWYPLYEYDAWGQGTLVTVSSGGGGS GGGSDVQLVESGGGLVQPGGSLRLSCAASGRTFSSYSMGWFRQAPGKEREFWAISKGGYKYDAVSL EGRFTISRDNAKNTVYLQINSLRPEDTAVYYCASSRAYGSSRLRLADTYEYWGQGTLVTVSSGGGGS GGGSEVQLVESGGGLVQPGNS LRLSCAASG FTFSS FGMSWVRQAPGKGLEWVS SISGSGSDT LYADS VKGRFTISRDNAKTTLYLQMNSLRPEDTAVYYCTIGGSLSRSSQGTLVTVSS (SEQ ID NO: 188)
VEGFANGBII00004 DVQLVESGGGLVQPGGSLRLSCAASGRTFSSYSMGWFRQAPGKEREFVVAISKGGYKYDAVSLEGRF TISRDNAKNTVYLQINSLRPEDTAVYYCASSRAYGSSRLRLADTYEYWGQGTLVTVSSAAAEVQLVE SGGGLVQPGNSLRLSCAASGFTFSSFGMSWVRQAPGKGLEWVSSISGSGSDTLYADSVKGRFTISRD NAKTTLYLQMNSLRPEDTAVYYCTIGGSLSRSSQGTLVTVSSAAAEVQLVESGGGLVQAGGSLRLSC AASGFTLDDYAIGWFRQAPGKEREGVSSIRDNDGSTYYADSVKGRFTISSDNDKNTVYLQMNSLKPE DTAVYYCAAVPAGRLRFGEQWYPLYEYDAWGQGTLVTVSS (SEQ ID NO: 189)
125
Table 38-B
VHH ID
Format
Expression (mg/L)
VEGFANGBII00001
13.3
VEGFANGBII00002
11.3
VEGFANGBII00003
OttTT
DOOM
15.4
VEGFANGBII00004
19,2
To explore the anti-VEGF blocking properties in comparison with the monovalent building block VEGFBII00038, ail four bispecific VHHs are analyzed in the VEGF/VEGFR2-Fc (Figure 22) compétition AlphaScreen. The assay is siightly adjusted compared to Example 12.3 described in patent US 2011/0172398 A1. Both human VEGF165 and human VEGFR2-Fc are added at 0.05 nM. This compétition assay is also performed after preincubation of the VHH with 25 μΜ human sérum albumin. A summary of IC5o values and % inhibition is shown in Table 39.
Table 39: IC50 values (nM) in human VEGF165/human VEGFR2 compétition AlphaScreen
VHH ID
Format
HSA
ICao (nM)
VEGFBII00038
VEGFANGBII00001
VEGFANGBII00002
V w
0,5
100
VEGFANGBII00003
0.5
Ô4
100
100
0.6
0.7
100
100
100
100
126
+ 1.3 100
VEGFANGBII00004 ^^3 yLBty ^^3 - 0.5 100
+ 0.7 100
Ranibizumab - 0.8 100
+ 1.1 100
To explore the anti-Ang2 blocking properties in comparison with the monovalent building block 00027 (SEQ ID No:216), ail four bispecific VHHs are analyzed in a human Ang2/hTie2-Fc (Figure 25) compétition ELISA. This assay is also performed after incubation of the VHH with 0,5 pM human sérum albumin. A summary of IC50 values is shown in Table 40.
Table 40: IC50 values (pM) in human Ang2/human Tie2 compétition ELISA
VHH ID
Format
HSA
IC50 (pM)
00027
00077
516
n.d.
VEGFANGBI100001
00038
240
179
VEGFANGBI I00002
00038
463
330
VEGFANGBI I00003
273
VEGFANGBI I00004
AMG386
n.d., not determined
00038
154
111
n.d.
127
Example 11
Construction, production and characterization of trivalent and tetravalent bispecific VHHs targeting VEGF and Ang2 using anti-serum albumin binding as half-life extension
Ten bispecific VHHs targeting VEGF and Ang2 are constructed (VEGFANGBI100005-00015). In these constructs monovalent and bivalent 1D01 (SEQ ID NO:214), monovalent and bivalent 7G08 (SEQ ID NO:215) and bivalent 00027 (SEQ ID NO:216) anti-Ang2 building blocks are included. A genetic fusion to a sérum albumin binding VHH is used as half-life extension methodology. Building blocks are linked via a 9 Gly-Ser flexible linker. VHHs are produced and purified as described in Example 8. An overview of the format and sequence of ail ten bispecific VHHs is depicted in Figure 26 and Table 41-A (linker sequences underlined), SEQ ID Nos 190-199. Expression levels are indicated in Table 41-B.
Table 41-A
Sequences of bispecific VHH targeting VEGF and Ang2
VHH ID AA sequence
VEGFANGBII00005 DVQLVESGGGLVQPGGSLRLSCAASGRTFSSYSMGWFRQAPGKEREFWAISKGGYKYDAVSLEGRF TISRDNAKNTVYLQINSLRPEDTAVYYCASSRAYGSSRLRLADTYEYWGQGTLVTVSSGGGGSGGGS EVQLVESGGGLVQPGGSLRLSCAASGFALDYYAIGWFRQVPGKEREGVSCISSSDGITYYVDSVKGR FTISRDNAKNTVYLQMNSLKPE DTAVYYCAT DSGGYIDYDCMGLGYDYWGQGTLVTVS SGGGGSGGG SEVQLVESGGGLVQPGNSLRLSCAASGFTFSSFGMSWVRQAPGKGLEWVSSISGSGSDTLYADSVKG RFTISRDNAKTTLYLQMNSLRPEDTAVYYCTIGGSLSRSSQGTLVTVSS (SEQ ID NO: 190)
VEGFANGBII00006 DVQLVESGGGLVQPGGSLRLSCAASGRTFSSYSMGWFRQAPGKEREFVVAISKGGYKYDAVSLEGRF TISRDNAKNTVYLOINSLRPEDTAVYYCASSRAYGSSRLRLADTYEYWGQGTLVTVSSGGGGSGGGS EVQLVESGGGLVQPGNS LRLS CAASG FT FSS FGMSWVRQAPGKGLEWVSSISGSGS DT LYADSVKGR FTISRDNAKTTLYLQMNSLRPEDTAVYYCTIGGSLSRSSQGTLVTVSSGGGGSGGGSEVQLVESGGG LVQPGGSLRLSCAASGFALDYYAIGWFRQVPGKEREGVSCISSSDGITYYVDSVKGRFTISRDNAKN TVYLQMNSLKPEDTAVYYCATDSGGYIDYDCMGLGYDYWGQGTLVTVSS (SEQ ID NO: 191)
VEGFANGBII00007 DVQLVESGGGLVQPGGSLRLSCAASGRTFSSYSMGWFRQAPGKEREFWAISKGGYKYDAVSLEGRF TISRDNAKNTVYLQINSLRPEDTAVYYCASSRAYGSSRLRLADTYEYWGQGTLVTVSSGGGGSGGGS EVQLVESGGGLVQPGGSLRLSCAASGFALDYYAIGWFRQVPGKEREGVSCISSSDGITYYVDSVKGR FTISRDNAKNTVYLQMN SLK PEDTAVYYCATDSGGYIDYDCMGLGYDYWGQGTLVTVS SGGGGSGGG SEVQLVESGGGLVQPGNSLRLSCAASGFTFSSFGMSWVRQAPGKGLEWVSSISGSGSDTLYADSVKG RFTISRDNAKTTLYLQMNS LRPE DTAVYYCTIGGS LSRSSQGT LVTVS SGGGGSGGGSEVQLVE SGG GLVQPGGSLRLSCAASGFALDYYAIGWFRQVPGKEREGVSCISSSDGITYYVDSVKGRFTISRDNAK NTVYLQMNSLKPEDTAVYYCATDSGGYIDYDCMGLGYDYWGQGTLVTVSS (SEQ ID NO: 192)
VEGFANGBI100008 DVQLVESGGGLVQPGGSLRLSCAASGRTFSSYSMGWFRQAPGKEREFWAISKGGYKYDAVSLEGRF TISRDNAKNTVYLQINSLRPEDTAVYYCASSRAYGSSRLRLADTYEYWGQGTLVTVSSGGGGSGGGS EVQLVESGGGLVQAGGSLRLSCAASGFTLDDYAIGWFRQAPGKEREGVSSIRDNDGSTYYADSVKGR FTISSDNDKNTVYLQMNSLKPEDTAVYYCAAVPAGRLRFGEQWYPLYEYDAWGQGTLVTVSSGGGGS GGGSEVQLVESGGGLVQAGGSLRLSCAASGFTLDDYAIGWFRQAPGKEREGVSSIRDNDGSTYYADS VKGRFTISSDNDKNTVYLQMNSLKPEDTAVYYCAAVPAGRLRFGEQWYPLYEYDAWGQGTLVTVSSG GGGSGGGSEVQLVESGGGLVQPGNSLRLSCAASGFT FSSFGMSWVRQAPGKGLEWVS SISGSGS DTL
128
YADSVKGRFTISRDNAKTTLYLQMNSLRPEDTAVYYCTIGGSLSRSSQGTLVTVSS (SEQ ID NO: 193)
VEGFANGBI100009 DVQLVESGGGLVQPGGSLRLSCAASGRTFSSYSMGWFRQAPGKEREFWAISKGGYKYDAVSLEGRF TISR DNAKNTVYLQINSLR PE DTAVYYCAS SRAYGS SRLRLADTYEYWGQGT LVTVSSGGGGSGGGS EVQLVESGGGLVQPGNSLRLSCAASGFTFSSFGMSWVRQAPGKGLEWVSSISGSGSDTLYADSVKGR FTISRDNAKTTLYLQMNSLRPEDTAVYYCTIGGSLSRSSQGTLVTVSSGGGGSGGGSEVQLVESGGG LVQAGGSLRLSCAASGFTLDDYAIGWFRQAPGKEREGVSSIRDNDGSTYYADSVKGRFTISSDNDKN TVYLQMNSLKPEDTAVYYCAAVPAGRLRFGEQWYPLYEYDAWGQGTLVTVSSGGGGSGGGSEVQLVE SGGGLVQAGGSLRLSCAASGFTLDDYAIGWFRQAPGKEREGVSSIRDNDGSTYYADSVKGRFTISSD NDKNTVYLQMNSLKPEDTAVYYCAAVPAGRLRFGEQWYPLYEYDAWGQGTLVTVSS (SEQ ID NO: 194)
VEGFANGBII00010 DVQLVESGGGLVQPGGSLRLSCAASGRTFSSYSMGWFRQAPGKEREFWAISKGGYKYDAVSLEGRF TISRDNAKNTVYLQINSLRPEDTAVYYCASSRAYGSSRLRLADTYEYWGQGTLVTVSSGGGGSGGGS EVQLVESGGGLVQAGGSLRLSCAASGFTLDDYAIGWFRQAPGKEREGVSSIRDNDGSTYYADSVKGR FTISSDNDKNTVYLQMNSLKPEDTAVYYCAAVPAGRLRFGEQWYPLYEYDAWGQGTLVTVSSGGGGS GGGSEVQLVESGGGLVQPGNSLRLSCAASGFTFSSFGMSWVRQAPGKGLEWVSSISGSGSDTLYADS VKGRFTISRDNAKTTLYLQMNS LRPEDTAVYYCTIGGSLSRS SQGTLVTVS SGGGGSGGGSEVQLVE SGGGLVQAGGSLRLSCAASGFTLDDYAIGWFRQAPGKEREGVSSIRDNDGSTYYADSVKGRFTISSD NDKNTVYLQMNSLKPEDTAVYYCAAVPAGRLRFGEQWYPLYEYDAWGQGTLVTVSS (SEQ ID NO: 195)
VEGFANGBIIÛ0011 DVQLVESGGGLVQPGGSLRLSCAASGRTFSSYSMGWFRQAPGKEREFVVAISKGGYKYDAVSLEGRF TISRDNAKNTVYLQINSLRPEDTAVYYCASSRAYGSSRLRLADTYEYWGQGTLVTVSSGGGGSGGGS EVQLVESGGGLVQAGGSLRLSCAASGFTFDDYALGWFRQAAGKEREGVSCIRCSDGSTYYADSVKGR FTIS S DNAKNTVYLQMNS LKPE DTAVYYCAASIVPRSKLE PYEY DAWGQGT LVTVSSGGGGSGGGS E VQLVESGGGLVQAGGSLRLSCAASGFTFDDYALGWFRQAAGKEREGVSCIRCSDGSTYYADSVKGRF TISSDNAKNTVYLQMNSLKPEDTAVYYCAASIVPRSKLEPYEYDAWGQGTLVTVSSGGGGSGGGSEV QLVESGGGLVQ PGNSLRLS CAASGFT FS S FGMSWVRQAPGKGLEWVS SISGSGSDTLYADSVKGR FT ISRDNAKTTLYLQMNSLRPEDTAVYYCTIGGSLSRSSQGTLVTVSS (SEQ ID NO: 196)
VEGFANGBII00012 DVQLVESGGGLVQPGGSLRLSCAASGRTFSSYSMGWFRQAPGKEREFWAISKGGYKYDAVSLEGRF TISRDNAKNTVYLQINSLRPEDTAVYYCASSRAYGSSRLRLADTYEYWGQGTLVTVSSGGGGSGGGS EVQLVESGGGLVQPGNSLRLSCAASGFTFSSFGMSWVRQAPGKGLEWVSSISGSGSDTLYADSVKGR FTIS R DNAKTTLYLQMNSLRPE DTAVYYCTIGGS LSRSSQGTLVTVS SGGGGSGGGSEVQLVESGGG LVQAGGSLRLSCAASGFTFDDYALGWFRQAAGKEREGVSCIRCSDGSTYYADSVKGRFTISSDNAKN TVYLQMNSLKPEDTAVYYCAASIVPRSKLEPYEYDAWGQGTLVTVSSGGGGSGGGSEVQLVESGGGL VQAGGSLRLSCAASGFTFDDYALGWFRQAAGKEREGVSCIRCSDGSTYYADSVKGRFTISSDNAKNT VYLQMNSLKPEDTAVYYCAASIVPRSKLEPYEYDAWGQGTLVTVSS (SEQ ID NO: 197}
VEGFANGBI100013 DVQLVESGGGLVQPGGSLRLSCAASGRTFSSYSMGWFRQAPGKEREFWAISKGGYKYDAVSLEGRF TISRDNAKNTVYLQINSLRPEDTAVYYCASSRAYGSSRLRLADTYEYWGQGTLVTVSSGGGGSGGGS EVQLVESGGGLVQAGGSLRLSCAASGFTFDDYALGWFRQAAGKEREGVSCIRCSDGSTYYADSVKGR FTISSDNAKNTVYLQMNSLKPEDTAVYYCAASIVPRSKLEPYEYDAWGQGTLVTVSSGGGGSGGGSE VQLVESGGGLVQPGNSLRLSCAASGFTFSSFGMSWVRQAPGKGLEWVSSISGSGSDTLYADSVKGRF TISRDNAKTTLYLQMNSLRPEDTAVYYCTIGGSLSRSSQGTLVTVSSGGGGSGGGSEVQLVESGGGL VQAGGSLRLSCAASGFTFDDYALGWFRQAAGKEREGVSCIRCSDGSTYYADSVKGRFTISSDNAKNT VYLQMNSLKPEDTAVYYCAASIVPRSKLEPYEYDAWGQGTLVTVSS (SEQ ID NO: 198)
VEGFANGBI100014 DVQLVESGGGLVQPGGSLRLSCAASGRTFSSYSMGWFRQAPGKEREFWAISKGGYKYDAVSLEGRF TISRDNAKNTVYLQINSLRPEDTAVYYCASSRAYGSSRLRLADTYEYWGQGTLVTVSSGGGGSGGGS EVQLVESGGGLVQPGN SLRLSCAASGFT FS S FGMSWVRQAPGKGLEWVS SISGSGSDTLYADSVKGR FTIS RDNAKTTLYLQMNS LRPE DTAVYYCTIGGS LSRS SQGTLVTVS SGGGGSGGGSEVQLVESGGG LVQAGGSLRLSCAASGFTFDDYALGWFRQAAGKEREGVSCIRCSDGSTYYADSVKGRFTISSDNAKN TVYLQMNSLKPEDTAVYYCAASIVPRSKLEPYEYDAWGQGTLVTVSS (SEQ ID NO: 199)
Table 41-B
VHH ID Format Expression (mg/L)
VEGFANGBII00005 1.3
VEGFANGBII00006 1.3
129
VEGFANGBII00007
VEGFANGBII00008
VEGFANGBII00009
VEGFANGBII00010
0.3
30.0
71.4
25.0
VEGFANGBII00011
VEGFANGBII00012
VEGFANGBII00013
VEGFANGBII00014
00038
00038
00038
00038
4.0
ΡΡρφ~φ>
ΠΡΡΦ
6.6
2.9
45.6
Το explore the anti-VEGF blocking properties in comparison with the monovalent building block VEGFBII00038, ail ten bispecific VHHs are analyzed in the VEGF/VEGFR2-Fc (Example 10; Figure 27-1) and VEGF/VEGFR1 (Figure 27-2) compétition AlphaScreen. The VEGFR1 assay is slightly adjusted compared to Example 12.4 as described in patent US 2011/0172398 A1. Human VEGF165 and human VEGFR1-Fc are added at 0.05 nM. These compétition assays are also performed after preincubation of the VHH with 25 pM human sérum albumin. A summary of IC5o values is shown in Table 42.
Table 42: IC50 values (nM) in human VEGF165/human VEGFR2 and human
VEGF165/human VEGFR1 compétition AlphaScreen
VEGFR1
VEGFR2
VHH ID
Format
HSA
IC50 (nM)
VEGFBII00038
00038
0.6
0.6
IC50 (nM)
0.5
0.5 % inh
100
100
VEGFANGBII00005
n.d.
n.d.
n.d.
n.d.
n.d.
n.d.
n.d.
n.d.
VEGFANGBII00006
0.8
1.0
100
1.0
1.4
100
130
VEGFANGBII00007
MÛ38
VEGFANGBII00008
VEGFANGBII00009
00038
VEGFANGBI100010
VEGFANGBI100001
VEGFANGBII00011
00038
VEGFANGBII00012
0Û038
VEGFANGBII00013
00038
VEGFANGBII00014
Ranibizumab
n.d., not determined
To explore the anti-Ang2 blocking properties in comparison with their respective monovalent building block 7G08 (SEQ ID No:215), 1D01 (SEQ ID No:214) and 00027 (SEQ ID No:216), ail ten bispecific VHHs are analyzed in the human Ang2/hTie2-Fc (see Example 5.1; Figure 28-1), mouse Ang2/mTie2-Fc (see Example 5.2; Figure 28-2) and cyno Ang2/cTie2-Fc (see Example 5.2; Figure 283) compétition ELISA. The human assay is also performed after incubation of the VHH with 0.5 μΜ human sérum albumin. Additionally, a hAng2 mediated HUVEC survival assay is performed (see Example 5.5; Figure 29). A summary of IC50 values and % inhibition is shown in Table 43.
131
Table 43: IC50 values (pM) and % inhibition in human Ang2/human Tîe2, mouse Ang2/mouse Tie2 and cyno Ang2/cyno Tie2 > e n » o LU > ZD
TJ Φ ro
TJ Φ E
CM CO C < x:
c
HUVEC survival % inh 100 n.d. xj c n.d. 100 n.d. n.d. TJ c 100 n.d. 100 TJ c 100 TJ c
O J in n.d. pu n.d. 4.9 n.d. n.d. n.d. 1.2 n.d. 2.0 c 5.7 n.d.
CM CT C S % inh n.d. XJ c n.d. n.d. xi c TJ c TJ c n.d. n.d. TJ c 100 TJ c 100 n.d.
sS' o CT n.d. n.d. n.d. n.d. n.d. n.d. n.d. n.d. n.d. pu 570 n.d. 390 n.d.
CM CT C < E % inh n.d. pu 1 xj c TJ c: tj c TJ c TJ c TJ c TJ c TJ c 100 n.d. 100 n.d.
8 Ç - J n.d. tj c n.d. n.d. n.d. n.d. n.d. n.d. TJ c n.d. 1,800 n.d. 1,300 n.d. |
CM CT C < -C % inh 100 pu n.d. ; n.d. 100 100 n.d. n.d. 100 100 100 n.d. 100 100
o J o TJ c n.d. n.d. 560 400 i nd- n.d. m 540 n.d. 360 290
HSA + 1 + 1 + i + 1 1 + 1 +
Format < 1 J o i
VHH ID 7G08 VEGFANGBII 00005 VEGFANGBII 00006 VEGFANGBII 00007 AMG386 00027 VEGFANGBII 00001
132
100 n.d. 100 n.d. b c b c 100 n.d. 100 n.d. 100 n.d. 100 n.d. n.d. b c n.d. n.d. n.d.
5.6. •b c 3.6. Ό C n.d. n.d. cq V“ pu 7.8 n.d. 3.7 n.d. 3.6 pu n.d. n.d. n.d. n.d. 1.3 n.d.
100 ri c 100 n.d. 100 n.d. o o Ύ- q c o o T“ Ό C 100 n.d. o o pu pu n.d. 100 n.d. 100 n.d.
79 n.d. 46 n.d. 59 n.d. co n.d. 9,500 n.d. 95 n.d. 100 pu pu n.d o o n.d O T n.d.
100 Ό c 100 n.d. o o n.d 100 n.d. o o pu 100 n.d. 100 pu b c n.d. 100 n.d. 100 n.d.
n.d. 36 n.d. 78 n.d. n.d. 000'01. n.d. 56 n.d. 89 n.d. pu n.d. o o o n.d. M- n.d.
100 O O r— 100 100 100 100 100 100 100 n.d. 100 100 100 100 n.d. n.d. | 100 100 100 100
47 52 33 39 32 49 m 7,000 pu co 34 40 65 •b c •b b 710 1,200 -d- 't
1 + 1 4- + + 1 + + 1 + 1 + 4- 4-
0 ' £ : 1 I t
II ni fi i |I| II II
VEGFANGBII 00008 VEGFANGBII 00009 VEGFANGBII 00010 AMG386 1D01 VEGFANGBII 00011 VEGFANGBII 00012 VEGFANGBII 00013 VEGFANGBII 00014 AMG386
n.d., not determined
133
Affinities of for human sérum albumin hâve been determined and are shown in Table 44. Briefly, human sérum albumin (Sigma, St Louis, MO, USA) is immobilized on a CM5 chip via amine coupling. A multicycle kinetic approach is used: increasing concentrations of VHH (2-8-31-125-500 nM) are injected and allowed to associate for 2 min and to dissociate for 10 min at a flow rate of 100 pL/min. Between VHH injections, the surfaces are regenerated with a 10 sec puise of 10 mM Glycine-HCI pH 1.5 and 60 sec stabilization period. Association/dissociation data are evaluated by fitting a 1:1 interaction model (Langmuir binding) or Heterogeneous Ligand model. The affinity constant Ko is calculated from resulting association and dissociation rate constants ka and kd (Table 44).
134
Table 44: Affinity KD of purified VHHs for human (HSA), cyno (CSA) and mouse sérum albumin (MSA)
MSA o B X c 49 Ό C pu 360 n.d. n.d. 340 n.d. •pu 240 n.d. n.d.
•D 3.2E-02 pu T) c 6.0E-02 pu pu 4.0E-02 n.d. pu 2.5E-02 n.d. n.d.
*u? m *> 6.6E+05 n.d. pu 1.7E+05 n.d. n.d. 1.2E+05 n.d. pu 1.0E+05 xi c pu
CSA Q Ξ * S 24 n.d. 30 n.d. pu 39 n.d. n.d. 24 q c: n.d.
1.6E-03 4.3E-03 n.d. 4.5E-03 n.d. pu 4.2E-03 n.d. pu 4.2E-03 6 c n.d.
(Λ* § 4.3E+05 1.8E+05 pu 1.5E+05 TJ c pu 1.1E+05 pu pu 1.2E+05 •pu TJ c
HSA Q X c 22 n.d. 22 xi G 34 30 •pu eo T* co q c n.d.
T. λ B 1 1,8E-03 I 4.8E-03 pu 4.6E-03 n.d. 4.3E-03 4.6E-03 pu 4.0E-03 I 4.3E-03 pu n.d.
i to o + LU to 2.3E+05 pu 2.0E+05 q c 1.3E+05 1.5E+05 Ό C 1.3E+05 1.5E+05 pu Ό c
ALB11 VEGFANGBII00001 VEGFANGBII00005 VEGFANGBII00006 VEGFANGBII00007 VEGFANGBII00008 VEGFANGBII00009 VEGFANGBII00010 VEGFANGBII00011 VEGFANGBII00012 CO t— o 2 CD O z £ 0 UJ > VEGFANGBII0014
n.d., not determined
135
Example 12
Construction, production and characterization of sequence optimized and affinity matured bispecific VHHs targeting VEGF and Ang2 using anti-serum albumin binding as half-life extension bispecific VHHs targeting VEGF and Ang2 are constructed (VEGFANGBII00015-00028). In these constructs bivalent 00921 (a sequence optimized 1D01 variant) (SEQ ID No:220), monovalent VHHs 00908 - 00932 00933 - 00934 - 00935 - 00936 - 00937 - 00938 (sequence optimized/affinity matured 28D10 variants) (SEQ ID No:222), bivalent 00956 (SEQ ID NO:223) (sequence optimized 28D10 variant) and monovalent 00928 (SEQ ID NO:221) (sequence optimized 37F02 variant) anti-Ang2 building blocks are included. A genetic fusion to a sérum albumin binding VHH is used as half-life extension methodology. Building blocks are linked via a 9 Gly-Ser flexible linker. An overview of the format and sequence of al) 14 bispecific VHHs is depicted in Figure 30 and Table 45-A (linker sequences underlined), SEQ ID Nos 200-213. Expression levels are indicated in Table 45-B.
Table 45-A
Sequences of bispecific. VHH targeting VEGF and Ang2
VHH ID AA sequence
VEGFANGBI100015 DVQLVES GGGLVQPGGS LRLSCAAS GRT FSSYSMGWFRQAPGKERE FWAISKGGYKYDAVSLEGRF TISRDNAKNTVYLQINSLRPEDTAVYYCASSRAYGSSRLRLADTYEYWGQGTLVTVSSGGGGSGGGS EVQLVESGGGLVQPGNS LRL S CAASG FT FSS FGMSWVRQAPGKGLEWVSSISGSGS DT LYAD SVKGR FTISRDNAKTTLYLQMNSLRPEDTAVYYCTIGGSLSRSSQGTLVTVSSGGGGSGGGSEVQLVESGGG LVQPGGS LRL S CAASGITLD DYAIGW FRQAPGKEREGVSSIRDNGGSTYYADSVKGRFTISS DNS KN TVYLQMNSLRPEDTAVYYCAAVPAGRLRYGEQWYPIYEYDAWGQGTLVTVSS (SEQ ID NO: 200)
VEGFANGBI100016 DVQLVESGGGLVQPGGSLRLSCAASGRTFSSYSMGWFRQAPGKEREFWAISKGGYKYDAVSLEGRF TISRDNAKNTVYLQINSLRPEDTAVYYCASSRAYGSSRLRLADTYEYWGQGTLVTVSSGGGGSGGGS EVQLVESGGGLVQPGNS LRLSCAASG FT FSS FGMSWVRQAPGKGLEWVSSISGSGS DT LYADSVKGR FTISRDNAKTTLYLQMNSLRPEDTAVYYCTIGGSLSRSSQGTLVTVSSGGGGSGGGSEVQLVESGGG LVQPGGS LRLS CAVSGITLD DYAIGW FRQAPGKEREGVSSIRDNGGSTYYADSVKGRFTISS DN SKN TVYLQMNSLRPEDTAVYYCAAVPAGRLRYGEQWYPIYEYDAWGQGTLVTVSS SEQ ID NO: 201)
VEGFANGBI100017 DVQLVESGGGLVQPGGSLRLSCAASGRTFSSYSMGWFRQAPGKEREFWAISKGGYKYDAVSLEGRF TISRDNAKNTVYLQINSLRPEDTAVYYCASSRAYGSSRLRLADTYEYWGQGTLVTVSSGGGGSGGGS EVQLVESGGGLVQPGNS LRLSCAASGFT FSS FGMSWVRQAPGKG LEWVSSISGSGS DT LYADSVKGR FTISRDNAKTTLYLQMNSLRPEDTAVYYCTIGGSLSRSSQGTLVTVSSGGGGSGGGSEVQLVESGGG LVQPGGSLRLSCAASGITLDDYAIGWFRQAPGKEREGVSAIRDNGGSTYYADSVKGRFTISSDNSKN TVYLQMNSLRPEDTAVYYCAAVPAGRLRYGEQWYPIYEYDAWGQGTLVTVSS SEQ ID NO: 202)
136
VEGFANGBII00018 DVQLVESGGGLVQPGGSLRLSCAASGRTFSSYSMGWFRQAPGKEREFWAISKGGYKYDAVSLEGRF TISRDNAKNTVYLQINSLRPEDTAVYYCASSRAYGSSRLRLADTYEYWGQGTLVTVSSGGGGSGGGS EVQLVE SGGGLVQPGNS LRL S CAASG FT FSS FGMSWVRQAPGKGLEWVS SISG SG SDTLYADSVKGR FTISRDNAKTTLYLOMNSLRPEDTAVYYCTIGGSLSRSSQGTLVTVSSGGGGSGGGSEVQLVESGGG LVQ PGGSLRLSCAAS GITLD DYAIGW FRQAPGKEREGVSAIRE SGGSTYYADSVKGRFTISS DN SKN TVYLQMNSLRPEDTAVYYCAAVPAGRLRYGEQWYPIYEYDAWGQGTLVTVSS SEQ ID NO: 203)
VEGFANGBII00019 DVQLVESGGGLVQPGGSLRLSCAASGRTFSSYSMGWFRQAPGKEREFWAISKGGYKYDAVSLEGRF TISRDNAKNTVYLQINSLRPEDTAVYYCASSRAYGSSRLRLADTYEYWGQGTLVTVSSGGGGSGGGS EVQLVESGGGLVQPGNSLRLSCAASGFTFSSFGMSWVRQAPGKGLEWVSSISGSGSDTLYADSVKGR FTISRDNAKTTLYLQMNSLRPEDTAVYYCTIGGSLSRSSQGTLVTVSSGGGGSGGGSEVQLVESGGG LVQ PGGSLRLSCAASGITLDDYAIGW FRQAPGKEREGVSAIRS SGGSTYYADSVKGRFTISS DN SKN TVYLQMNSLRPEDTAVYYCAAVPAGRLRYGEQWYPIYEYDAWGQGTLVTVSS SEQ ID NO: 204)
VEGFANGBII00020 DVQLVESGGGLVQPGGSLRLSCAASGRTFSSYSMGWFRQAPGKEREFWAISKGGYKYDAVSLEGRF TISRDNAKNTVYLOINSLRPEDTAVYYCASSRAYGSSRLRLADTYEYWGQGTLVTVSSGGGGSGGGS EVQLVE SGGG LVQPGN SLRLSCAASG FT FSS FGMSWVRQAPGKGLEWVSSISGS GS DTLYADSVKGR FTISRDNAKTTLYLQMNSLRPEDTAVYYCTIGGSLSRSSQGTLVTVSSGGGGSGGGSEVQLVESGGG LVQPGGSLRLSCAVSGITLDDYAIGWFRQAPGKEREGVSAIRDNGGSTYYADSVKGRFTISSDNSKN TVYLQMNSLRPEDTAVYYCAAVPAGRLRYGEQWYPIYEYDAWGQGTLVTVSS SEQ ID NO: 205)
VEGFANGBII00021 DVQLVESGGGLVQPGGSLRLSCAASGRTFSSYSMGWFRQAPGKÊREFWAISKGGYKYDAVSLEGRF TISRDNAKNTVYLQINSLRPEDTAVYYCASSRAYGSSRLRLADTYEYWGQGTLVTVSSGGGGSGGGS EVQLVES GGGLVQPGN S LRLSCAASG FT FSS FGMSWVRQAPGKGLEWVSSISGSGS DT LYADSVKGR FTISRDNAKTTLYLQMNSLRPEDTAVYYCTIGGSLSRSSQGTLVTVSSGGGGSGGGSEVQLVESGGG LVQPGGSLRLS CAVSGITLDDYAIGW FRQAPGKEREGVSAIRE SGGSTYYADSVKGR FTISS DN SKN TVYLQMNSLRPEDTAVYYCAAVPAGRLRYGEQWYPIYEYDAWGQGTLVTVSS SEQ ID NO: 206)
VEGFANGBII00022 DVQLVESGGGLVQPGGSLRLSCAASGRTFSSYSMGWFRQAPGKEREFWAISKGGYKYDAVSLEGRF TISRDNAKNTVYLQINSLRPEDTAVYYCASSRAYGSSRLRLADTYEYWGQGTLVTVSSGGGGSGGGS EVQLVE SGGGLVQPGN SLRL SCAASG FT FSS FGMSWVRQAPGKGLEWVS SISG SGS DTLYADSVKGR FTISRDNAKTTLYLQMNSLRPEDTAVYYCTIGGSLSRSSQGTLVTVSSGGGGSGGGSEVQLVESGGG LVQ PGGSLRLSCAVSGIT LD DYAIGWFRQAPGKEREGVSAIRS SGGSTYYADSVKGRFTISS DN SKN TVYLQMNSLRPEDTAVYYCAAVPAGRLRYGEQWYPIYEYDAWGQGTLVTVSS SEQ ID NO: 207)
VEGFANGBII00023 DVQLVESGGGLVQPGGSLRLSCAASGRTFSSYSMGWFRQAPGKEREFWAISKGGYKYDAVSLEGRF TISRDNAKNTVYLQINSLRPEDTAVYYCASSRAYGSSRLRLADTYEYWGQGTLVTVSSGGGGSGGGS EVQLVESGGGLVQPGNSLRLSCAASGFTFSSFGMSWVRQAPGKGLEWVSSISGSGSDTLYADSVKGR FTISRDNAKTTLYLQMNSLRPEDTAVYYCTIGGSLSRSSQGTLVTVSSGGGGSGGGSEVQLVESGGG LVQPGGSLRLSCAASGFTLDDYAIGWFRQAPGKEREGVSAIRDNGGSTYYADSVKGRFTISSDNSKN TVYLQMNSLRPEDTAVYYCAAVPAGRLRFGEQWYPLYEYDAWGQGTLVTVSSGGGGSGGGSEVQLVE SGGGLVQ PGGSLRLS CAAS GFTLDDYAIGW FRQAPGKEREGVSAIRDNGGSTYYADSVKGRFTISS D NSKNTVYLQMNSLRPEDTAVYYCAAVPAGRLRFGEQWYPLYEYDAWGQGTLVTVSS SEQ ID NO: 208}
VEGFANGBII00024 DVQLVE SGGGLVQPGG S LRLSCAASGRT FS S YSMGW FRQAPGKEREFWAISKGGYKYDAVS LEGRF TISRDNAKNTVYLQINSLRPEDTAVYYCASSRAYGSSRLRLADTYEYWGQGTLVTVSSGGGGSGGGS EVQLVE SGGGLVQPGGSLRLSCAASG FTLD DYAIGW FRQAPGKEREGVSAIRDNGGSTYYADSVKGR FTISSDNSKNTVYLQMNSLRPEDTAVYYCAAVPAGRLRFGEQWYPLYEYDAWGQGTLVTVSSGGGGS GGGSEVQLVESGGGLVQPGGSLRLSCAASGFTLDDYAIGWFRQAPGKEREGVSAIRDNGGSTYYADS VKGRFTISSDNSKNTVYLQMNSLRPEDTAVYYCAAVPAGRLRFGEQWYPLYEYDAWGQGTLVTVSSG GGGSGGGSEVQLVESGGGLVQPGNSLRLSCAASGFTFSSFGMSWVRQAPGKGLEWVSSISGSGSDTL YADSVKGRFTISRDNAKTTLYLQMNSLRPEDTAVYYCTIGGSLSRSSQGTLVTVSS SEQ ID NO: 209)
VEGFANGBII00025 DVQLVESGGGLVQPGGS LRLS CAASGRT FSSYSMGW FRQAPGKEREFWAISKGGYKYDAVS LEGRF TISRDNAKNTVYLQINSLRPEDTAVYYCASSRAYGSSRLRLADTYEYWGQGTLVTVSSGGGGSGGGS EVQLVESGGGLVQPGNSLRLSCAASGFTFSSFGMSWVRQAPGKGLEWVSSISGSGSDTLYADSVKGR FTISRDNAKTTLYLQMNSLRPEDTAVYYCTIGGSLSRSSQGTLVTVSSGGGGSGGGSEVQLVESGGG LVQPGGSLRLSCAASGFTFDDYALGWFRQAPGKEREGVSCIRCSGGSTYYADSVKGRFTISSDNSKN TVYLQMNSLRPEDTAVYYCAASIVPRSKLEPYEYDAWGQGTLVTVSSGGGGSGGGSEVQLVESGGGL VQPGGSLRLSCAASGFTFDDYALGWFRQAPGKEREGVSCIRCSGGSTYYADSVKGRFTISSDNSKNT VYLQMNSLRPEDTAVYYCAASIVPRSKLEPYEYDAWGQGTLVTVSS SEQ ID NO: 210}
137
VEGFANGBII00026 DVQLVESGGGLVQPGGSLRLSCAASGRTFSSYSMGWFRQAPGKEREFWAISKGGYKYDAVSLEGRF TISRDNAKNTVYLQINSLRPEDTAVYYCASSRAYGSSRLRLADTYEYWGQGTLVTVSSGGGGSGGGS EVQLVESGGGLVQPGGSLRLSCAASGFTFDDYALGWFRQAPGKEREGVSCIRCSGGSTYYADSVKGR FTISSDNSKNTVYLQMNSLRPEDTAVYYCAASIVPRSKLEPYEYDAWGQGTLVTVSSGGGGSGGGSE VQLVESGGGLVQPGGSLRLSCAASGFTFDDYALGWFRQAPGKEREGVSCIRCSGGSTYYADSVKGRF TISSDNSKNTVYLQMNSLRPEDTAVYYCAASIVPRSKLEPYEYDAWGQGTLVTVSSGGGGSGGGSEV QLVESGGGLVQPGNSLRLSCAASGFTFSSFGMSWVRQAPGKGLEWVSSISGSGSDTLYADSVKGRFT ISRDNAKTTLYLQMNSLRPEDTAVYYCTIGGSLSRSSQGTLVTVSS SEQ ID NO: 211)
VEGFANGBII00027 DVQLVESGGGLVQPGGSLRLSCAASGRTFSSYSMGWFRQAPGKEREFWAISKGGYKYDAVSLEGRF TISRDNAKNTVYLQINSLRPEDTAVYYCASSRAYGSSRLRLADTYEYWGQGTLVTVSSGGGGSGGGS EVQLVESGGGLVQPGNSLRLSCAASGFTFSSFGMSWVRQAPGKGLEWVSSISGSGSDTLYADSVKGR FTISRDNAKTTLYLQMNSLRPEDTAVYYCTIGGSLSRSSQGTLVTVSSGGGGSGGGSEVQLVESGGG LVQ PGGSLRLSCAASGFALDYYAIGW FRQAPGKEREGVSCISS SGGITYYADSVKGRFTISRDNSKN TVYLQMNSLRPEDTAVYYCATDSGGYIDYDCSGLGYDYWGQGTLVTVSS SEQ ID NO: 212)
VEGFANGBII00028 DVQLVESGGGLVQPGGSLRLSCAASGRTFSSYSMGWFRQAPGKEREFWAISKGGYKYDAVSLEGRF TISRDNAKNTVYLQINSLRPEDTAVYYCASSRAYGSSRLRLADTYEYWGQGTLVTVSSGGGGSGGGS EVQLVESGGGLVQPGNSLRLSCAASGFTFSSFGMSWVRQAPGKGLEWVSSISGSGSDTLYADSVKGR FTISRDNAKTTLYLQMNSLRPEDTAVYYCTIGGSLSRSSQGTLVTVSSGGGGSGGGSEVQLVESGGG LVQPGGSLRLSCAASGFTLDDYAIGWFRQAPGKEREGVSAIRSSGGSTYYADSVKGRFTISSDNSKN TVYLQMNSLRPEDTAVYYCAAVPAGRLRFGEQWYPLYEYDAWGQGTLVTVSSGGGGSGGGSEVQLVE SGGGLVQPGGSLRLS CAASG FTLDDYAIGWFRQAPGKEREGVSAIRS S GGSTYYADSVKGR FTISS D NSKNTVYLQMNSLRPEDTAVYYCAAVPAGRLRFGEQWYPLYEYDAWGQGTLVTVSS SEQ ID NO: 213)
Table 45-B
VHH ID Format Expression (mg/L)
VEGFANGBII00022 00038 [ WBiy
VEGFANGBII00025 00038
VEGFANGBI100028 00030 W w w
To explore the anti-VEGF blocking properties in comparison with the monovalent building block VEGFBII00038, the bispecific VHHs are analyzed in the VEGF/VEGFR2-Fc (Example 10; Figure 31 -1 ) and VEGF/VEGFR1 (Example 11 ; Figure 31-2) compétition AlphaScreen. These compétition assays are also performed after preincubation of the VHH with 25 μΜ human sérum albumin. A summary of IC50 values is shown in Table 46-A.
138
Table 46-A IC50 values (nM) in human VEGF165/human VEGFR2 and human
VEGF165/human VEGFR1 compétition AlphaScreen
VEGFR1
VEGFR2
VHH ID
Format
HSA
IC50 (nM)
iCgo (nM) % inh
VEGFBII00038
Ranïbizumab
VEGFANGBII00022
VEGFANGBII00025
VEGFANGBII00028
00038
00021
00038
0Û15S £ OOD5S
0.4
n.d.
0.4
0.6
0.6
0.9
0.5
0.5
3.2
n.d.
0.2
100
n.d.
n.d.
n.d.
n.d.
0.2
0.3
0.2
0.3
0.2
0.2
0.7
0.9
100
100
100
100
100
100
100
100
n.d., not determined
Binding kinetics of the bispecific VHHs on human VEGF165 is analyzed by SPR on a Biacore T100 instrument (see Example 12.5 described in patent US 2011/0172398 A1). Monovalent Nanobody VEGFBII00038 is taken along as référencé (Table 46-B).
Table 46-B: Overview of kinetic parameters in hVEGF165 Biacore assay.
ka1 (1/Ms) kd1 (1/s) ka2 (1/s) kd2 (1/s) KD1 (M)
VEGFBII00038 2.6E+05 1.3E-02 1.3E-02 1.9E-04 7.5E-10
VEGFANGBII00022 1.6E+05 1.4E-02 1.4E-02 2.2E-04 1.4E-09
VEGFANGBII00025 1.1E+05 1.4E-02 1.4E-02 2.1E-04 1.9E-09
VEGFANGBII00028 1.7E+05 1.3E-02 1.3E-02 2.1E-04 1.1E-09
139
The ability of the VHHs to bind to human isoform VEGF121 is determined in a binding ELISA. Binding of a dilution sériés of VHH to 1 pg/mL directly coated human VEGF121 (R&D) (human VEGF165 as référencé) is detected using biotinylated anti-VHH 1A4 followed by extravidin-HRP. 1A4 is a anti-VHH VHH (generated in-house by Ablynx NV). The benchmark Avastin serves as positive control and is detected using a H RP conjugated anti-human Fc antibody. An irrelevant VHH serves as négative control. Représentative binding response curves on VEGF165 and VEGF121 are shown in Figure 46 corresponding EC50 values are summarized in Table 46-C.
Table 46-C: Overview of EC5o values in hVEGF165 and hVEGF121 binding ELISA.
hVEGF165 hVEGF121
EC50(/W) ECsq (M)
VEGFANGBII00022 1.4E-09 2.3E-09
VEGFANGBII00025 1.5E-09 2.5E-09
VEGFANGBII00028 1.2E-09 2.1E-09
Binding to rat and mouse VEGF164 is assessed in a binding ELISA. VHHs binding to 1 pg/mL directly coated murine or rat VEGF164 (R&D) are detected using biotinylated anti-VHH 1A4 followed by extravidin-HRP. As positive control the mouse/rat cross-reactive monoclonal antibody B20-4.1 (Genentech) is titrated and detected with an HRP conjugated anti-human Fc antibody. An irrelevant VHH serves as négative control. Results are shown in Figure 33. Ail 3 bispecific VHH are not cross-reactive to mouse and rat VEGF.
140
Binding to human VEGF-B, VEGF-C, VEGF-D and PIGF is assessed via a binding ELISA. Binding of VHHs to 1 pg/mL directly coated VEGF-B (R&D), VEGF-C (R&D), VEGF-D (R&D) and PIGF (R&D) was detected using biotinylated anti-VHH 1 A4 followed by extravidin-HRP. As positive controls a sériés of dilutions of the appropriate receptors (hVEGFR1-Fc for hVEGF-B and hPIGF, hVEGFR2-Fc for hVEGF-C, anti-hVEGF-D mAb (R&D) for hVEGF-D) are taken along. An irrelevant VHH serves as négative control. Results are shown in Figure 34. Ail 3 bispecific VHH are not binding to VEGF family members.
To explore the anti-Ang2 blocking properties in comparison with their respective monovalent building block 00921 (SEQ ID NO:220) and 00938 (SEQ ID NO:222), ail 3 bispecific VHHs are analyzed in the human Ang2/hTie2-Fc (see Example 5.1; Figure 35-1), mouse Ang2/mTie2-Fc (see Example 5.2; Figure 35-2) and cyno Ang2/cTie2-Fc (see Example 5.2; Figure 35-3) compétition ELISA. The human assay is also performed after incubation of the VHH with 0.5 pM human sérum albumin. Additionally, bispecific VHHs are tested in the hAng1/hTie2 compétition ELISA (see Example 5.3; Figure 36) and the Ang2 mediated HUVEC survival assay (see Example 5.5; Figure 37). A summary of IC5o values and % inhibition is shown in Table 47-A.
141
Table 47-A: IC5o values (pM) in human Ang2/human Tie2, mouse Ang2/mouse Tie2 and cyno Ang2/cyno Tie2 compétition
ELISA, hAngl ELISA and IC50 values (nM) in hAng2 mediated HUVEC survival assay
HUVEC survival % inh o o 100 pu 100 o o T— o o pu 100 100 100 pu 100
IC50 (nM) 4.3 1.9 pu 1.4 18.8 2.2 n.d. 1.2 6.8 3.7 pu o
ELISA CD C < -C CM o c S ç c - < jz > 60,395 > 39,902 > 44,771 5,656 > 125 > 160,694 > 133,660 5,632 > 1,979 > 66,222 >50,816 5,194
CM CJ> c s % inh 100 100 100 100 100 100 o o 100 100 100 100 100
Sî ο a *0»< 86 o 115 co 43,500 38 37 0 1,294 65 78 TT
CM CD C < E % inh 100 100 100 100 100 100 100 100 100 100 100 100
8s ç 1 58 56 68 CM 27,990 lo V“ r-- CM 1,816 72 69 co
Ang2 % inh 100 100 100 100 100 100 100 100 100 100 o o 100
Ça 33 50 45 0 15,940 CM V 0 0 1,010 30 39 M*
HSA 1 + 1 1 1 + t t + 1
Format e ? û ► ► 0 1
VHH ID 00938 CM CM O O O ώ O Z < LL ω LU > AMG386 00921 VEGFANGBII00025 AMG386 00956 VEGFANGBII00028 AMG386
n.d., not determined
142
Affinities of VEGFANGBII00022-25-28 for human, mouse, cyno and rat Ang2 (see
Example 5.4) hâve been determined and are shown in Table 47-B.
Table 47-B: Affinity KD of purified VHHs for recombïnant human, cyno, mouse and rat Ang2
human Ang2-FLD cyno Ang2-FLD
ka (1/Ms) kd (1/s) KD (M) ka (1/Ms) kd (1/s) Kd (M)
VEGFANGBII00022 9.7E+05 1.5E-05 1.6E-11 1.5E+06 1.3E-05 8.1 E-12
VEGFANGBII00025 2.7E+06 1.2E-02 4.5E-09 4.3E+06 1.1E-02 2.7E-09
VEGFANGBII00028 5.9E+05 9.6E-04 1.6E-09 8.4E+05 8.7E-04 1.0E-09
mouse Ang2-FLD rat Ang2-FLD
ka (1/Ms) kd (1/s) Kd (M) ka (1/Ms) kd (1/s) KD (M)
VEGFANGBII00022 5.5E+05 2.8E-05 5.1E-11 3.9E+05 3.8E-05 9.9E-11
VEGFANGBII00025 1.3E+06 1.4E-02 1.1E-08 8.7E+05 2.9E-02 3.3E-08
VEGFANGBII00028 3.6E+05 2.0E-03 5.6E-09 2.5E+05 3.1E-03 1.2E-08
Affinities of VEGFANGBH00022-25-28 for human, mouse and cyno sérum albumin hâve been determined (Example 11 ) and are shown in Table 48. The affinity constant w Kd is calculated from resulting association and dissociation rate constants ka and kd (Table 48).
143
Table 48: Affinity KD (nM) of purified VHHs for recombinant human, mouse and cyno sérum albumin using (A) 1:1 interaction model or (B) heterogeneous ligand model
(A) HSA CSA
ka (1/Ms) kd (1/s) Kq (nM) ka (1/Ms) kd (1/s) Kd (nM)
ALB11 5.6E+05 1.9E-03 4 4.5E+05 1.7E-03 4
VEGFANGBII00022 6.7E+05 6.0E-03 9 6.2E+05 5.4E-03 9
VEGFANGBII00025 5.6E+05 5.6E-03 12 4.3E+05 5.1E-03 12
VEGFANGBII00028 5.6E+05 5.8E-03 10 5.2E+05 5.3E-03 10
MSA
ka (1/Ms) kd (1/s) Ko (nM)
ALB11 5.9E+05 3.0E-02 51
VEGFANGBII00022 5.2E+05 5.4E-03 150
VEGFANGBII00025 - - -
VEGFANGBII00028 - - -
(B) MSA
ka1 (1/Ms) kdi (1/s) ka2 (1/s) kd2 (1/s). Kdi (nM) Kü2 (nM)
VEGFANGBII00025 6.2E+05 9.9E-02 4.7E+04 5.7E-04 160* 12
VEGFANGBII00028 5.9E+04 6.9E-04 5.7E+05 9.4E-02 12 160*
* describes 70% or more of the interaction
144
Ang2-binding components (Table 49) (1D01 (SEQ ID No: 214); 7G08 (SEQ ID No:215); 027 (SEQ ID No:216); 00042 (SEQ ID No:217); 00050 (SEQ DI No:218); 00045 (SEQ ID No:219); 00921 (SEQ ID No:220); 00928 (SEQ ID No:221); 00938 (SEQ ID No:222); 00956 (SEQ ID No:223)
1D01
7G08
027
EVQLVESGGGLVQAGGSLRLSCAASGFTFD
EVQLVESGGGLVQPGGSLRLSCAASGFALD
EVQLVESGGGLVQAGGSLRLSCAASGFTLD
DYALG
YYAIG
DYAIG
WFRQAAGKEREGVS WFRQVPGKEREGVS
WFRQAPGKEREGVS
CIRCSDGSTYYADSX
CISSSDGITYYVDS\
CIRDSDGSTYYADSX
1D01
7G08
027
SIVPRSKLEPYEYDA
DSGGYIDYDCMGLGYDY
VPAGRLRFGEQWYPLYEYDA
WGQGTQVTVSS
WGQGTQVTVSS
WGQGTQVTVSS
00042
00050
00045 o»
00042
00050
00045
DYAIG
DYALG
YYAIG
WFRQAPGKEREGVS
WFRQAPGKEREGVS
WFRQAPGKEREGVS &·: ‘ï·
VPAGRLRFGEQWYPLYEYDA SIVPRSKLEPYEYDA DSGGYIDYDCMGLGYDY
SIRDNDGSTYYADSV1
CIRCSDGSTYYADSV1
CISSSDGITYYADSV1
WGQGTLVTVSS WGQGTLVTVSS WGQGTLVTVSS
00921
00928
00938
00956
00921
00928
00938
00956
EVQLVESGGGLVQPGGSLRLSCAASGFTFD DYALG EVQLVESGGGLVQPGGSLRLSCAASGFALD YYAIG EVQLVESGGGLVQPGGSLRLSCAVSGÏTLD DYAIG EVQLVESGGGLVQPGGSLRLSCAASGFTLD DYAIG
RFTISSDNSKNTVYLQMNSLRPEDTAVYYCAA RFTISRDNSKNTVYLQMNSLRPEDTAVYYCAT RFTISSDNSKNTVYLQMNSLRPEDTAVYYCAA RFTISSDNSKNTVYLQMNSLRPEDTAVYYCAA
WFRQAPGKEREGVS WFRQAPGKEREGVS WFRQAPGKEREGVS WFRQAPGKEREGVS •*jfî*jmirwitKTnw
CIRCSGGSTYYADSV CISSSGGITYYADSV AIRSSGGSTYYADSV AIRSSGGSTYYADSV
SIVPRSKLEPYEYDA DSGGYIDYDCSGLGYDY VPAGRLRYGEQWYPIYEYDA VPAGRLRFGEQWYPLYEYDA
WGQGTLVTVSS WGQGTLVTVSS WGQGTLVTVSS WGQGTLVTVSS

Claims (39)

1. A bispecific binding molécule comprising at least one VEGF-binding component, at least one Ang2-binding component and at least one sérum albumin binding component, wherein said Ang2-binding component binds to Ang2 with a potency at least 5,000 times higher than to Ang1 or to Ang4.
2. A bispecific binding molécule of claim 1, wherein said VEGF-binding component comprises at least a variable domain with four framework régions and three complementarity determining régions CDR1, CDR2 and CDR3, respectively, wherein said CDR3 has the amino acid sequence Ser Arg Ala Tyr Xaa Ser Xaa Arg Leu Arg Leu Xaa Xaa Thr Tyr Xaa Tyr as shown in SEQ ID NO: 1, wherein
Xaa at position 5 is Gly or Ala;
Xaa at position 7 is Ser or Gly;
Xaa at position 12 is Gly, Ala or Pro;
Xaa at position 13 is Asp or Gly;
Xaa at position 16 is Asp or Glu; and wherein said VEGF-binding component is capable of blocking the interaction of human recombinant VEGF165 with the human recombinant VEGFR-2 with an inhibition rate of >60%.
3. A bispecific binding molécule of claim 2, wherein said CDR3 has a sequence selected from
SEQ ID NO: 2 SRAYGSSRLRLGDTYDY, SEQ ID NO: 3 SRAYGSSRLRLADTYDY; SEQ ID NO: 4 SRAYGSSRLRLADTYEY; SEQ ID NO: 5 SRAYGSGRLRLADTYDY; SEQ ID NO: 6 SRAYASSRLRLADTYDY; SEQ ID NO: 7 SRAYGSSRLRLPDTYDY; SEQ ID NO: 8 SRAYGSSRLRLPGTYDY.
146
4. A bispecific binding molécule of claim 3, wherein said VEGF-binding component comprises one or more immunoglobulin single variable domains each containing
a. a CDR3 with an amino acid sequence selected from a first group of sequences shown in SEQ ID NO: 2 to 8;
b. a CDR1 and a CDR2 with an amino acid sequences that is contained, as indicated in Table 3, in a sequence selected from a second group of sequences shown in SEQ ID NOs: 9 to 46, wherein said second sequence contains the respective CDR3 in said selected sequence according to a).
5. A bispecific binding molécule of claim 4, wherein said one or more immunoglobulin single variable domains are VHHs.
6. A bispecific binding molécule of claim 5, wherein said one or more VHHs hâve amino acid sequences selected from the amino acid sequences shown in SEQ ID NOs: 9 - 46.
7. A bispecific binding molécule of claim 6, which comprises one or more VHHs having amino acid sequences selected from SEQ ID NO: 15, SEQ ID NO: 18 and SEQ ID NO: 25.
8. A bispecific binding molécule, the VEGF-binding component of which has been obtained by affinity maturation and/or sequence optimization of a VHH defined in claim 7.
9. A bispecific binding molécule according to claim 8, the VEGF-binding component of which has been obtained by sequence optimization of a VHH having an amino acid sequence shown in SEQ ID NO: 18.
147
10. A bispecific binding molécule according to claim 9, the VEGF-binding component of which having an amino acid sequence selected from sequences shown in SEQ ID NOs: 47 - 57.
11. A bispecific binding molécule according to claim 5, the VEGF-binding component of which comprising two or more VHHs, which are
a. identical VHHs that are capable of blocking the interaction between recombinant human VEGF and the recombinant human VEGFR-2 with an inhibition rate of à 60% or
b. different VHHs that bind to non-overlapping epitopes of VEGF, wherein at least one VHH is capable of blocking the interaction between recombinant human VEGF and the recombinant human VEGFR-2 with an inhibition rate of s 60% and wherein at least one VHH is capable of blocking said interaction with an inhibition rate of s 60 %.
12. A bispecific binding molécule according to claim 11, wherein said identical VHHs a) are selected from VHHs having amino acid sequences shown in SEQ ID NOs: 9 - 46 or VHHs that hâve been obtained by affinity maturation and/or sequence optimization of such VHH.
13. A bispecific binding molécule according to claim 12, wherein said VHH is selected from VHHs having the amino acid shown in SEQ ID NO: 18 or SEQ IDNO: 47-57.
14. A bispecific binding molécule according to claim 13 comprising two VHHs each having the amino acid sequence shown in SEQ ID NO: 57.
15. A bispecific binding molécule according to claim 14, wherein
a. said one or more VHHs with an inhibition rate of z 60% are selected from
148
i. VHHs having an amino acid sequence selected from amino acid sequences shown in SEQ ID NOs: 9 - 46 or ii. VHHs that hâve been obtained by affinity maturation and/or sequence optimization of such VHHs, and wherein
b. said one or more VHHs with an inhibition rate of ê 60 % are selected from
i. SEQ ID NOs: 58-124 or ii. VHHs that hâve been obtained by affinity maturation and/or sequence optimization of such VHH.
16. A bispecific binding molécule according to claim 15, wherein two VHHs are contained in polypeptides with amino acid sequences shown in SEQ ID NOs: 128 - 168, separated by linker sequences as indicated in Table 13.
17. A bispecific binding molécule according to claim 16, wherein said VHH a) i. has an amino acid sequence shown in SEQ ID NO: 18 and said VHH b) i. has an amino acid sequence shown in SEQ ID NO: 64.
18. A bispecific binding molécule according to claim 17, wherein said VHHs according to a) ii) are selected from VHHs having an amino acid sequence shown in SEQ ID NOs: 47 - 57 and wherein said VHHs according to b) ii) are selected from VHHs having an amino acid sequence shown in SEQ ID NOs: 125-127.
19. A bispecific binding molécule according to claim 18, comprising two VHHs, one of them having the amino acid shown in SEQ ID NO: 57 and one of them having the amino acid shown in SEQ ID NO: 127.
20 selected from a second group of amino acid sequences shown SEQ ID
NOs: 224, 227, 230, 233, 236, 239, 242, 245, 248, or 251 (Table 49);
c. a CDR2 with an amino acid sequences that is contained, as indicated in
Table 36-A, 38-A, 41-A, or 45-A, as partial sequence in a sequence selected from a second group of amino acid sequences shown SEQ ID
NOs:225, 228, 231,234, 237, 240, 243, 246, 249, or 252 (Table 49).
20. The bispecific binding molécule of any one of daims 1 to 19, comprising an Ang2-binding component comprising at least a variable domain with four
149 framework régions and three complementarity determining régions CDR1, CDR2 and CDR3, respectively, wherein said CDR3 has an amino acid sequence selected from amino acid sequences shown in SEQ IDs NOs: 226, 229, 232, 235, 238, 241, 244, 247, 250, or 253.
s
21. The bispecific binding molécule of claim 20, the Ang2-binding component of which is an isolated immunoglobulin single variable domain or a polypeptide containing one or more of said immunoglobulin single variable domains, wherein said immunoglobulin single variable domain consists of four framework régions and three complementarity determining régions CDR1, w CDR2 and CDR3, respectively, and wherein said CDR3 has an amino acid sequence selected from amino acid sequences shown in SEQ IDs NOs: 226, 229, 232, 235, 238, 241,244, 247, 250, or 253.
22. The bispecific binding molécule of claim 21, wherein said one or more immunoglobulin single variable domain contain
a. a CDR3 with an amino acid sequence selected from a first group of amino acid sequences shown in SEQ ID NOs: SEQ IDs NOs: 226,
229, 232, 235, 238, 241,244, 247, 250, or 253 (Table 49);
b. a CDR1 with an amino acid sequences that is contained, as indicated in
Table 36-A, 38-A, 41-A, or 45-A, as partial sequence in a sequence
23. The bispecific binding molécule of any one of daims 20 to 22, wherein said one or more immunoglobulin single variable domains are VHHs.
150
24. The bispecific binding molécule of claim 23, wherein said one or more VHHs hâve an amino acid sequence selected from amino acid sequences shown in SEQ ID NOs: 214, 215, 216, 217, 218, 219, 220, 221, 222, or 223.
25. An immunoglobulin single variable domain which has been obtained by affinity maturation of an immunoglobulin single variable domain as defined in claim 22.
26. A VHH which has been obtained by affinity maturation of a VHH as defined in claim 24.
27. An Ang2-binding VHH with an amino acid sequence selected from acid sequences shown in SEQ ID NOs: 214, 215, 216, 217, 218, 219, 220, 221, 222, or 223.
28. An immunoglobulin single variable domain which has been obtained by humanization of a VHH defined in claim 27.
29. An immunoglobulin single variable domain which has been obtained by humanization of an immunoglobulin single variable domain as defined in claim 22.
30. The binding molécule of any one of claim 1 to 29, the sérum albumin binding component of which is an isolated immunoglobulin single variable domain or a polypeptide containing one or more of said immunoglobulin single variable domains, wherein said immunoglobulin single variable domain consists of four framework régions and three complementarity determining régions CDR1, CDR2 and CDR3, respectively, and wherein said CDR3 has an amino acid sequence selected from amino acid sequences shown in SEQ ID NOs: 257, 260, 263, 266, 269, 272, or 275.
31. The binding molécule of claim 30, wherein said one or more immunoglobulin single variable domain contain
151
a. a CDR3 with an amino acid sequence selected from a first group of amino acid sequences shown in SEQ ID NOs: SEQ IDs NOs: 257, 260, 263, 266, 269, 272, or 275;
b. a CDR1 with an amino acid sequences selected from a second group of amino acid sequences shown SEQ ID NOs: 255, 258, 261,264, 267,
270, or 273;
c. a CDR2 with an amino acid sequences selected from a second group of amino acid sequences shown SEQ ID NOs: 256, 259, 262, 265, 268,
271, or 274.
32. The bispecific binding molécule of claim 30 or 31, wherein said one or more immunoglobulin single variable domains are VHHs.
33. The bispecific binding molécule of claim 32, wherein said one or more VHHs hâve an amino acid sequence shown in SEQ ID NOs: 98 or 254.
34. The bispecific binding molécule of any one of daims 1 to 33 having the amino acid sequence selected from amino acid sequences shown in SEQ ID NOs: 180-213.
35. A nucleic acid molécule encoding a bispecific binding molécule of any one of daims 1 to 34 or a vector containing same.
36. A host cell containing a nucleic acid molécule of claim 35.
37. A pharmaceutical composition containing at least one bispecific binding molécule of any one of daims 1 to 34 as the active ingrédient.
38. The pharmaceutical composition of claim 37 for the treatment of a disease that is associated with VEGF- and/or Ang2-mediated effects on angiogenesis.
152
39.
s 40.
The pharmaceutical composition of claim 38 for the treatment of cancer and cancerous diseases.
The pharmaceutical composition of claim 38 for the treatment of eye diseases.
OA1201300405 2011-04-01 2012-03-30 Bispecific molecules binging to VEGF and Ang2. OA16601A (en)

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
EP11160921.0 2011-04-01

Publications (1)

Publication Number Publication Date
OA16601A true OA16601A (en) 2015-11-20

Family

ID=

Similar Documents

Publication Publication Date Title
US20220017642A1 (en) Bispecific binding molecules binding to VEGF and Ang2
US20220363744A1 (en) Vegf-binding molecules
CA2865464C (en) Ang2-binding molecules
US20110195494A1 (en) Dll4-binging molecules
WO2011039370A1 (en) Bispecific binding molecules for anti-angiogenesis therapy
US20130078247A1 (en) Bispecific binding molecules binding to dii4 and ang2
OA16601A (en) Bispecific molecules binging to VEGF and Ang2.
OA16772A (en) Bispecific binding molecules binding to DII4 and Ang2.
NZ614249B2 (en) Bispecific binding molecules binding to vegf and ang2