Specific binding molecules for scintigraphy

Abstract
The present invention relates to antibodies with sub-nanomolar affinity specific for a characteristic epitope of the ED-B domain of fibronectin, a marker of angiogenesis. Furthermore, it relates to the use of radiolabeled high affinity anti ED-B antibodies for detecting new-forming blood vessels in vivo and a diagnostic kit comprising comprising said antibody.
Description


FIELD OF THE INVENTION

[0001] The present invention relates to antibodies with sub-nanomolar affinity specific for a characteristic epitope of the ED-B domain of fibronectin, a marker of angiogenesis. It also relates to the use of radiolabeled high-affinity anti-ED-B antibodies for detecting new-forming blood vessels in vivo and a diagnostic kit comprising said antibody.



BACKGROUND OF THE INVENTION

[0002] Tumours cannot grow beyond a certain mass without the formation of new blood vessels (angiogenesis), and a correlation between micro-vessel density and tumour invasiveness has been reported for a number of tumours (Folkman (1995). Nature Med., 1, 27-31). Molecules capable of selectively targeting markers of angiogenesis would create clinical opportunities for the diagnosis and therapy of tumours and other diseases characterised by vascular proliferation, such as diabetic retinopathy and age-related macular degeneration. Markers of angiogenesis are expressed in the majority of aggressive solid tumours and should be readily accessible to specific binders injected intravenously (Pasqualini et al. (1997). Nature Biotechnol., 15, 542-546; Neri et al. (1997), Nature Biotechnol., 15 1271-1275). Targeted occlusion of the neovasculature may result in tumour infarction and collapse (O'Reilly et al. (1996). Nature Med., 2, 689-692; Huang et al. (1997). Science, 275,- 547-550)


[0003] The ED-B domain of fibronectin, a sequence of 91 aminoacids identical in mouse, rat and human, which is inserted by alternative splicing into the fibronectin molecule, specifically accumulates around neo-vascular structures (Castellani et al. (1994). Int. J. Cancer 59, 612-618) and could represent a target for molecular intervention. Indeed, we have recently shown with fluorescent techniques that anti-ED-B single-chain Fv antibody fragments (scFv) accumulate selectively in tumoural blood vessels of tumour-bearing mice, and that antibody affinity appears to dictate targe-before, glycanase treatment renders these reagents unsuitable for applications according to the present invention. Recognition of ED-B(+)-FN in ELISA proceeds without the need of deglycosilation but only on cartilage extracted with a denaturing agent (4M Urea) and captured on plastic using gelatin. The authors comment that “the binding of the FN molecule to the gelatine bound on the plastic surface of the ELISA plate may somehow expose the epitopes sufficiently for recognition by the antiserum”. Since for in vivo applications FN cannot be denatured and gelatin bound, the monoclonal binders of the present invention offer distinct advantages.


[0004] The Japanese patents JP02076598 and JP04169195 refer to anti-ED-B antibodies. It is not clear from these documents if monoclonal anti ED-B antibodies are described. Moreover, it seems impossible that a single antibody (such as the antibody described in JP02076598) has “an antigen determinant in aminoacid sequence of formulae (1), (2) or (3):
1(1)EGIPIFEDFVDSSVGY(2)YTVTGLEPGIDYDIS(3)NGGESAPTTLTQQT


[0005] on the basis of the following evidence:


[0006] i) A monoclonal antibody should recognise a well-defined epitope.


[0007] ii) The three-dimensional structure of the ED-B domain of fibronectin has been determined by NMR spectroscopy. Segments (1), (2) and (3) lie on opposite faces of the ED-B structure, and cannot be bound simultaneously by one monoclonal antibody. Furthermore, in order to demonstrate the usefulness of the antibodies localisation in tumours should be demonstrated, as well as evidence of staining of ED-B(+)-FN structures in biological samples without treatment with structure-disrupting reagents.


[0008] The BC1 antibody described by Carnemolla et al. 1992, J. Biol. Chem. 267, 24689-24692, recognises an epitope on domain 7 of FN, but not on the ED-B domain, which is cryptic in the presence of the ED-B domain of fibronectin. It is strictly human-specific. Therefore, the BC1 antibody and the antibodies of the present invention show different reactivity. Furthermore, the BC1 antibody recognises domain 7 alone, and domain 7-8 of fibronectin in the absence of the ED-B domain (Carnemolla et al. 1992, J. Biol. Chem. 267, 24689-24692). Such epitopes could be produced in vivo by proteolytic degradation of FN molecules. The advantage of the reagents according to the present invention is that they can localise on FN molecules or fragments only if they contain the ED-B domain.


[0009] For the diagnosis of cancer, and more specifically for imaging primary and secondary tumour lesions, immunoscintigraphy is one of the techniques of choice. In this methodology, patients are imaged with a suitable device (e.g., a gamma camera), after having been injected with radiolabeled compound (e.g., a radionuclide linked to a suitable vehicle). For scintigraphic applications, short-lived gamma emitters such as technetium-99m, iodine-123 or indium-111 are typically used, in order to minimise exposure of the patient to ionising radiations.


[0010] The most frequently used radionuclide in Nuclear Medicine Departments is technetium-99m (99mTc), a gamma emitter with half-life of six hours. Patients injected with 99mTc-based radiopharmaceuticals can typically be imaged up to 12-24 hours after injections; however, accumulation of the nuclide on the lesion of interest at earlier time points is desirable.



SUMMARY OF THE INVENTION

[0011] Considering the need of nuclear medicine for radiopharmaceuticals capable of localising tumour lesions few hours after injection, and the information that antibody affinity appears to influence its performance in targeting of angiogenesis, it is an object of the present invention to produce antibodies specific for the ED-B domain of fibronectin with sub-nanomolar dissociation constant (for a review on the definitions and measurements of antibody-antigen affinity, see Neri et al. (1996). Trends in Biotechnol. 14, 465-470). A further object of the present invention is to provide radiolabeled antibodies in suitable format, directed against the ED-B domain of fibronectin, that detect tumour lesions already few hours after injection.


[0012] In one aspect of the invention these objects are achieved by an antibody with specific affinity for a characteristic epitope of the ED-B domain of fibronectin and with improved affinity to said ED-B epitope.


[0013] In a further aspect of the present invention the above described antibody is used for rapid targeting markers of angiogenesis.


[0014] Another aspect of the present invention is a diagnostic kit comprising said antibody and one or more reagents for detecting angiogenesis.


[0015] Still a further aspect of the present invention is the use of said antibody for diagnosis and therapy of tumours and diseases which are characterized by vascular proliferation.


[0016] Terminology


[0017] Throughout the application several technical expressios are used for which the following definitions apply.


[0018] antibody


[0019] This describes an immunoglobulin whether natural or partly or wholly synthetically produced. The term also covers any polypeptide or protein having a binding domain which is, or is homologous to, an antibody binding domain. These can be derived from natural sources, or they may be partly or wholly synthetically produced. Examples of antibodies are the immunoglobulin isotypes and their isotypic subclasses; fragments which comprise an antigen binding domain such as Fab, scFv, Fv, dAb, Fd; and diabodies. It is possible to take monoclonal and other antibodies and use techniques of recombinant DNA technology to produce other antibodies or chimeric molecules which retain the specificity of the original antibody. Such techniques may involve introducing DNA encoding the immunoglobulin variable region, or the complementarity determining regions (CDRs), of an antibody to the constant regions, or constant regions plus framework regions, of a different immunoglobulin. See, for istance, EP-A-184187, GB 2188638A or EP-A-239400. A hybridoma or other cell producing an antibody may be subject to genetic mutation or other changes, which may or may not alter the binding specificity of antibodies produced. As antibodies can be modified in a number of ways, the term “antibody” should be construed as covering any specific binding member or substance having a binding domain with the required specificity. Thus, this term covers antibody fragments, derivatives, functional equivalents and homologues of antibodies, including any polypeptide comprising an immunoglobulin binding domain, whether natural or wholly or partially synthetic. Chimeric molecules comprising an immunoglobulin binding domain, or equivalent, fused to another polypeptide are therefore included. Cloning and expression of chimeric antibodies are described in EP-A-0120694 and EP-A-0125023. It has been shown that fragments of a whole antibody can perform the function of binding antigens. Examples of binding fragments are (I) the Fab fragment consisting of VL, VH, CL and CH1 domains; (ii) the Fd fragment consisting of the VH and CH1 domains; (iii) the Fv fragment consisting of the VL and VH domains of a single antibody; (iv) the dAb fragment (Ward et al. (1989) Nature, ), 341, 544-546.) which consists of a VH domain; (v) isolated CDR regions; (vi) F(ab′)2 fragments, a bivalent fragment comprising two linked Fab fragments; (vii) single chain Fv molecules (scFv), wherein a VH domain and a VL domain are linked by a polypeptide linker which allows the two domains to associate to form an antigen binding site (Bird et al. (1988) Science, 242, 423-426.; Huston et al. (1988) Proc. Natl. Acad. Sci. U.S.A., 85, 5879-83.) ; (viii) bispecific single chain EV dimers (PCT/US92/09965) and (ix) “diabodies”, multivalent or multispecific fragments constructed by gene fusion (WO94/13804; Holliger et al. (1993) Proc. Natl. Acad. Sci. U.S.A., 90, 6444-6448). Diabodies are multimers of polypeptides, each polypeptide comprising a first domain comprising a binding region of an immunoglobulin light chain and a second domain comprising a binding region of an immunoglobulin heavy chain, the two domains being linked (e.g. by a petide linker) but unable to associate with each other to form an antigen binding site: antigen binding sites are formed by the association of the first domain of one polypeptide within the multimer with the second domain of another polypeptide within the multimer (WO94/13804). Where bispecific antibodies are to be used, these may be conventional bispecific antibodies, which can be manufactured in a variety of ways Holliger and Winter (1993), Curr. Opin. Biotech., 4, 446-449), e.g. prepared chemically or from hybrid hybridomas, or may be any of the bispecific antibody fragments mentioned above. It may be preferable to use scFv dimers or diabodies rather than whole antibodies. Diabodies and scFv can be constructed without an Fc region, using only variable domains, potentially reducing the effects of anti-idiotypic reaction. Other forms of bispecific antibodies include the single-chain CRAbs described by Neri et al. ((1995) J. Mol. Biol., 246,. 367-373).


[0020] complementarity-determining regions


[0021] Traditionally, complementarity-deterining regions (CDRs) of antibody variable domains have been identified as those hypervariable antibody sequences, containing residues essential for specific antigen recognition. In this document, we refer to the CDR definition and numbering of Chothia and Lesk (1987) J. Mol. Biol., 196, 901-917.


[0022] functionally equivalent variant form


[0023] This refers to a molecule (the variant) which altough having structural differences to another molecule (the parent) retains some significant homology and also at least some of the biological function of the parent molecule, e.g. the ability to bind a particular antigen or epitope. Variants may be in the form of fragments, derivatives or mutants. A variant, derivative or mutant may be obtained by modification of the parent molecule by the addition, deletion, substitution or insertion of one or more aminoacids, or by the linkage of another molecule. These changes may be made at the nucleotide or protein level. For example, the encoded polypeptide may be a Fab fragment which is then linked to an Fc tail from another source. Alternatively, a marker such as an enzyme, fluorescein, etc, may be linked. For example, a functionally equivalent variant form of an antibody “A” against a characteristic epitope of the ED-B domain of fibronectin could be an antibody “B” with different sequence of the complementarity determining regions, but recognising the same epitope of antibody “A”.


[0024] We have isolated recombinant antibodies in scFv format from an antibody phage display library, specific for the ED-B domain of fibronectin, and recognising ED-B(+)-fibronectin in tissue sections. One of these antibodies, E1, has been affinity matured to produce antibodies H10 and L19, with improved affinity. Antibody L19 has a dissociation constant for the ED-B domain of fibronectin in the sub-nanomolar concentration range.


[0025] The high-affinity antibody L19 and D1.3 (an antibody specific for an irrelevant antigen, hen egg lysozyme) were radiolabeled and injected in tumour-bearing mice. Tumour, blood and organ biodistributions were obtained at different time points, and expressed as percent of the injected dose per gram of tissue (% ID/g). Already 3 hours after injection, the % ID/g (tumour) was better than the % ID/g (blood) for L19, but not for the negative control D1.3. The tumour:blood ratios increased at longer time points. This suggests that the high-affinity antibody L19 may be a useful tumour targeting agent, for example for immunoscintigraphic detection of angiogenesis.







BRIEF DESCRIPTION OF THE DRAWINGS

[0026] Embodiments of the present invention are illustrated by the following figures, wherein


[0027]
FIG. 1 shows a designed antibody phage library;


[0028]
FIG. 2 shows 2D gels and Western blotting of a lysate of human melanoma COLO-38 cells;


[0029]
FIG. 3 shows immunohistochemical experiments of glioblastoma multiforme


[0030]
FIG. 4 shows an analysis of the stability of antibody-(ED-B) complexes.


[0031]
FIG. 5 shows biodistribution of tumour bearing mice injected with radiolabelled antibody fragments.


[0032]
FIG. 6 shows amino acid sequence of L19;


[0033]
FIG. 7 shows rabbit eyes with implanted pellet;


[0034]
FIG. 8 shows immunohistochemistry of rabbit cornea sections.







DETAILED DESCRIPTION OF THE DRAWINGS

[0035]
FIG. 1 shows:


[0036] Designed antibody phage library. (a) Antibody fragments are displayed on phage as pIII fusion, as schematically depicted. In the antibody binding site (antigen's eye view), the Vk CDRs backbone is in yellow, the VH CDR backbone is in blue. Residues subject to random mutation are Vk CDR3 positions 91, 93, 94 and 96 (yellow), and VH CDR3 positions 95, 96, 97, and 98 (blue). The Cb atoms of these side chains are shown in darker colours. Also shown (in gray), are the residues of CDR1 and CDR2, which can be mutated to improve antibody affinity. Using the program RasMol (http://www.chemistry.ucsc.edu/wipke/teaching/rasmol.html), the structure of the scFv were modeled from pdb file ligm (Brookhaven Protein Data Bank; http://www2.ebi.ac.uk/pcserv/pdbdb.htm). (b) PCR amplification and library cloning strategy. The DP47 and DPK22 germline templates were modified (see text) to generate mutations in the CDR3 regions. Genes are indicated as rectangles, and CDRs as numbered boxes within the rectangle. The VH and the VL segments were then assembled and cloned in pDN332 phagemid vector. Primers used in the amplification and assembly are listed at the bottom.


[0037]
FIG. 2) shows


[0038] 2D gels and western blotting (a) Silver-staining of the 2D-PAGE of a lysate of human melanoma COLO-38 cells, to which recombinant ED-B-containing 7B89 had been added. The two 7B89 spots (circle) are due to partial proteolysis of the His-tag used for protein purification. (b) Immunoblot of a gel, identical to the one of FIG. 2a, using the anti-ED-B E1 (Table 1) and the M2 anti-FLAG antibodies as detecting reagent. Only the 7B89 spots are detected, confirming the specificity of the recombinant antibody isolated from a gel spot.


[0039]
FIG. 3) shows:


[0040] Immunohistochemical experiments on serial sections of glioblastoma multiforme showing the typical glomerulus-like vascular structures stained using scFvs E1 (A), A2 (B) and G4 (C). Scale bars: 20 μm.


[0041]
FIG. 4) shows:


[0042] Stability of antibody-(ED-B) complexes. Analysis of the binding of scFvs E1, H10 and L19 to the ED-B domain of fibronectin. (a) BIA-core sensograms, showing the improved dissociation profiles obtained upon antibody affinity-maturation. (b) Native gel electrophoretic analysis of scFv-(ED-B) complexes. Only the high-affinity antibody L19 can form a stable complex with the fluorescently labeled antigen. Fluorescence detection was performed as described (Neri et al. (1996) BioTechniques, 20, 708-712). (c) Competition of the scFv-(ED-B-biotin) complex with a 100-fold molar excess of unbiotinylated ED-B, monitored by electrochemiluminescence using an Origen apparatus. A long half-life for the L19-(ED-B) complex can be observed. Black squares: L19; Open triangles: H10.


[0043]
FIG. 5) shows:


[0044] Biodistributions of tumour bearing mice injected with radiolabeled antibody fragments.


[0045] Tumour and blood biodistributions, expressed as percent injected dose per gram, are plotted versus time. Relevant organ biodistributions is also reported.


[0046]
FIG. 6) shows the amino acid sequence of antibody L19 comprising the heavy chain (VH), the linker and the light chain (VL).


[0047]
FIG. 7) shows rabbit eyes with implanted polymer pellets soaked with angiogenic substances.


[0048]
FIG. 8) shows Immunohistochemistry of sections of rabbit cornea with new-forming blood vessels, stained with the L19 antibody.


[0049] The invention is more closely discribed by the following examples.



EXAMPLE 1


Isolation of Human scFv Antibody Fragments Specific for the ED-B Domain of Fibronectin from a Antibody Phage-display Library

[0050] A human antibody library was cloned using VH (DP47; Tomlinson et al. (1992). J. Mol. Biol., 227 , 776-798.) and Vk (DPK22; Cox et al. (1994). Eur. J. Immunol., 24, 827-836) germline genes (see FIG. 1 for the cloning and amplification strategy). The VH component of the library was created using partially degenerated primers (FIG. 1) in a PCR-based method to introduce random mutations at positions 95-98 in CDR3. The VL component of the library was generated in the same manner, by the introduction of random mutations at positions 91, 93, 94 and 96 of CDR3. PCR reactions were performed as described (Marks et al. (1991). J. Mol. Biol., 222, 581-597). VH-VL scFv fragments were constructed by PCR assembly (FIG. 1; Clackson et al. (1991). Nature , 352, 624-628), from gel-purified VH and VL segments. 30 μg of purified VH-VL scFv fragments were double digested with 300 units each of NcoI and NotI, then ligated into 15 μg of Notl/Ncol digested pDN332 phagemid vector. pDN332 is a derivative of phagemid pHEN1 (Hoogenboom et al. (1991). Nucl. Acids Res., 19, 4133-4137), in which the sequence between the Notl site and the amber codon preceding the gene III has been replaced by the following sequence, coding for the D3SD3-FLAG-His6 tag (Neri et al. (1996). Nature Biotechnology, 14, 385-390):
2         Not1            D      D       D      S        D        DDY    K       D       D5′-GCG GCC GCA GAT GAC GAT TCC GAC GAT GAC TAC AAG GAC GAC   D        D       K       H      H    H      H        H        HamberGAC GAC AAG CAC CAT CAC CAT CAC CAT TAG-3′


[0051] Transformations into TGl E.coli strain were performed according to Marks et al. (1991. J. Mol. Biol., 222, 581-597) and phages were prepared according to standard protocols (Nissim et al. (1991). J. Mol. Biol., 222, 581-597). Five clones were selected at random and sequenced to check for the absence of pervasive contamination.


[0052] Recombinant fibronectin fragments ED-B and 7B89, containing one and four type III homology repeats respectively, were expressed from pQE12-based expression vectors (Qiagen, Chatsworth, Calif., USA) as described (Carnemolla et al. (1996). Int. J. Cancer, 68, 397-405).


[0053] Selections against recombinant ED-B domain of fibronectin (Carnemolla et al. (1996). Int. J. Cancer, 68, 397-405, Zardi et al. (1987). EMBO J., 6, 2337-2342) were performed at 10 nM concentration using the antigen biotinylated with biotin disulfide N-hydroxysuccinimide ester (reagent B-4531; Sigma, Buchs, Switzerland; 10) and eluted from a 2D gel, and streptavidin-coated Dynabeads capture (Dynal, Oslo, Norway). 1013 phages were used for each round of panning, in 1 ml reaction. Phages were incubated with antigen in 2% milk/PBS (MPBS) for 10 minutes. To this solution, 100 μl Dynabeads (10 mg/ml; Dynal, Oslo, Norway), preblocked in MPBS, were added. After 5 min. mixing, the beads were magnetically separated from solution and washed seven times with PBS-0.1% Tween-20 (PBST) and three times with PBS. Elution was carried out by incubation for 2 min. with 500 μl 50 mM dithiothreitol (DTT), to reduce the disulfide bridge between antigen and biotin. Beads were captured again, and the resulting solution was used to infect exponentially growing TG1 E.coli cells. After three rounds of panning, the eluted phage was used to infect exponentially-growing HB2151 E.coli cells and plated on (2xTY+1% glucose+100 μg/ml ampicillin)−1.5% agar plates. Single colonies were grown in 2xTY+0.1% glucose+100 μg/ml ampicillin, and induced overnight at 30 degrees with 1 mM IPTG to achieve antibody expression. The resulting supernatants were screened by ELISA using streptavidin-coated microtitre plates treated with 10 nM biotinylated-ED-B, and anti-FLAG M2 antibody (IBI Kodak, New Haven, Conn.) as detecting reagent. 32% of screened clones were positive in this assay and the three of them which gave the strongest ELISA signal (E1, A2 and G4) were sequenced and further characterised.


[0054] ELISA assays were performed using biotinylated ED-B recovered from a gel spot, biotinylated ED-B that had not been denatured, ED-B linked to adjacent fibronectin domains (recombinant protein containing the 7B89 domains), and a number of irrelevant antigens. Antibodies E1, A2 and G4 reacted strongly and specifically with all three ED-B containing proteins. This, together with the fact that the three recombinant antibodies could be purified from bacterial supernatants using an ED-B affinity column, strongly suggests that they recognise an epitope present in the native conformation of ED-B. No reaction was detected with fibronectin fragments which did not contain the ED-B domain (data not shown).


[0055] In order to test whether the antibodies isolated against a gel spot had a good affinity towards the native antigen, real-time interaction analysis was performed using surface plasmon resonance on a BIAcore instrument as described (Neri et al. (1997) Nature Biotechnol., 15, 1271-1275). Monomeric fractions of E1, A2 and G4 scFv fragments bound to ED-B with affinity in the 107-108 M-1 range (Table 1).


[0056] As a further test of antibody specificity and usefulness, a 2D-PAGE immunoblot was performed, running on gel a lysate of the human melanoma cell line COLO-38, to which minute amounts of the ED-B containing recombinant 7B89 protein had been added (FIG. 2). ScFv(E1) stained strongly and specifically only the 7B89 spot.


[0057] Antibodies E1, A2 and G4 were used to immunolocalise ED-B containing fibronectin (B-FN) in cryostat sections of glioblastoma multiforme, an aggressive human brain tumour with prominent angiogenetic processes. FIG. 3 shows serial sections of glioblastoma multiforme, with the typical glomerulus-like vascular structures stained in red by the three antibodies. Immunostaining of sections of glioblastoma multiforme samples frozen in liquid nitrogen immediately after removal by surgical procedures, was performed as described (Carnemolla et al. (1996). Int. J. Cancer, 68, 397-405, Castellani et al. (1994). Int. J. Cancer, 59, 612-618). In short, immunostaining was performed using M2-anti-FLAG antibody (IBI Kodak), biotinylated anti-mouse polyclonal antibodies (Sigma), a streptavidin-biotin alkaline phosphatase complex staining kit (BioSpa, Milan, Italy) and naphtol-AS-MX-phosphate and fast-red TR (Sigma). Gill's hematoxylin was used as a counter-stain, followed by mounting in glycergel (Dako, Carpenteria, Calif.) as previously reported (Castellani et al. (1994). Int. J. Cancer, 59, 612-618).


[0058] Using similar techniques and the antibody L19 (see next example) we could also specifically stain new-forming blood vessels induced by implanting in the rabbit cornea polymer pellets soaked with angiogenic substances, such as vascular endothelial growth factor or phorbol esters.



EXAMPLE 2


Isolation of a Human scFv Antibody Fragment Binding to the ED-B with Sub-nanomolar Affinity

[0059] ScFv(E1) was selected to test the possibility of improving its affinity with a limited number of mutations of CDR residues located at the periphery of the antigen binding site (FIG. 1A). We combinatorially mutated residues 31-33, 50, 52 and 54 of the antibody VH, and displayed the corresponding repertoire on filamentous phage. These residues are found to frequently contact the antigen in the known 3D-structures of antibody-antigen complexes. The resulting repertoire of 4×108 clones was selected for binding to the ED-B domain of fibronectin. After two rounds of panning, and screening of 96 individual clones, an antibody with 27-fold improved affinity was isolated (H10; Tables 1 and 2). Similarly to what others have observed with affinity-matured antibodies, the improved affinity was due to slower dissociation from the antigen, rather than by improved kon values (Schier et al. (1996). Gene, 169, 147-155, Ito (1995). J. Mol. Biol., 248, 729-732). The antibody light chain is often thought to contribute less to the antigen binding affinity as supported by the fact that both natural and artificial antibodies devoid of light chain can still bind to the antigen (Ward et al. (1989) Nature, 341, 544-546, Hamers-Casterman et al. (1993). Nature, 363, 446-448). For this reason we chose to randomise only two residues (32 and 50) of the VL domain,wich are centrally located in the antigen binding site (FIG. 1a) and often found in 3D structures to contact the antigen. The resulting library, containing 400 clones, was displayed on phage and selected for antigen binding. From analysis of the dissociation profiles using real-time interaction analysis with a BIAcore instrument (Jonsson et al. (1991). BioTechniques, 11, 620-627) and koff measurements by competition experiments with electrochemiluminescent detection a clone (L19) was identified, that bound to the ED-B domain of fibronectin with a Kd=54 pM (Tables 1 and 2).


[0060] Affinity maturation experiments were performed as follows. The gene of scFv(El) was PCR amplified with primers LMB1bis (5′-GCG GCC CAG CCG GCC ATG GCC GAG-3′) and DP47CDR1for (5′-GA GCC TGG CGG ACC CAG CTC ATM NNM NNM NNGCTA AAG GTG AAT CCA GAG GCT G-3′) to introduce random mutations at positions 31-33 in the CDR1 of the VH (for numbering: 28), and with primers DP47CDR1back (5′-ATG AGC TGG GTC CGC CAG GCT CC-3′) and DP47CDR2 for (5′-GTC TGC GTA GTA TGT GGT ACC MNN ACT ACC MNN AAT MNN TGA GAC CCA CTC CAG CCC CTT-3′) to randomly mutate positions 50, 52, 54 in CDR2 of the VH. The remaining fragment of the scFv gene, covering the 3′-portion of the VH gene, the peptide linker and the VL gene, was amplified with primers DP47CDR2back (5′-ACA TAC TAC GCA GAC TCC GTG AAG-3′) and JforNot (5′-TCA TTC TCG ACT TGC GGC CGC TTT GAT TTC CAC CTT GGT CCC TTG GCC GAA CG-3′) (94C 1 min, 60 C 1 min, 72 C 1 min). The three resulting PCR products were gel purified and assembled by PCR (21) with primers LMB1bis and JforNot (94° C. 1 min, 60 C 1 min, 72 C 1 min). The resulting single PCR product was purified from the PCR mix, double digested with NotI/NcoI and ligated into NotI/NcoI digested pDN332 vector. Approximately 9 μg of vector and 3 μg of insert were used in the ligation mix, which was purified by phenolisation and ethanol precipitation, resuspended in 50 μl of sterile water and electroporated in electrocompetent TGI E.coli cells. The resulting affinity maturation library contained 4×108 clones. Antibody-phage particles, produced as described (Nissim et al. (1994). EMBO J., 13, 692-698) were used for a first round of selection on 7B89 coated imunotube (Carnemolla et al. (1996). Int. J. Cancer, 68, 397-405). The selected phages were used for a second round of panning performed with biotinylated ED-B, followed by capture with streptavidin coated magnetic beads (Dynal, Oslo, Norway; see previous paragraph). After selection, approximately 25% of the clones were positive in soluble ELISA (see previous chapter for experimental protocol). From the candidates positive in ELISA, we further identified the one (H10; Table 1) with lowest koff by BIAcore analysis (Jonsson et al. (1991), BioTechniques, 11, 620-627).


[0061] The gene of scFv(H10) was PCR amplified with primers LMB1bis and DPKCDR1for (5′-G TTT CTG CTG GTA CCA GGC TAA MNN GCT GCT GCT AAC ACT CTG ACT G) to introduce a random mutation at position 32 in CDR1 of the VL (for numbering: 28), and with primers DPKCDR1back (5′-TTA GCC TGG TAC CAG CAG AAA CC-5′) and DPKCDR2for (5′-GCC AGT GGC CCT GCT GGA TGC MNN ATA GAT GAG GAG CCT GGG AGC C-3′) to introduce a random mutation at position 50 in CDR2 of the VL. The remaining portion of the scFv gene was amplified with oligos DPKCDR2back (5′-GCA TCC AGC AGG GCC ACT GGC-3′) and JforNot (94C 1 min, 60 C 1 min, 72 C 1 min) The three resulting products were assembled, digested and cloned into pDN332 as described above for the mutagenesis of the heavy chain. The resulting library was incubated with biotinylated ED-B in 3% BSA for 30 min., followed by capture on a streptavidin-coated microtitre plate (Boehringer Mannheim GmbH, Germany) for 10 minutes. The phages were eluted with a 20 mM DTT solution (1,4-Dithio-DL-threitol, Fluka) and used to infect exponentially growing TGl cells.


[0062] Analysis of ED-B binding of supernatants from 96 colonies by ELISA and by BIAcore allowed the identification of clone L19. Anti-ED-B E1, G4, A2, H10 and L19 scFv antibody fragments were produced inoculating a single fresh colony in 1 liter of 2xTY medium as previously described in Pini et al. ((1997), J. Immunol. Meth., 206, 171-182) and affinity purified onto a CNBr-activated sepharose column (Pharmacia, Uppsala, Sweden), which had been coupled with 10 mg of ED-B containing 7B89 recombinant protein (Carnemolla et al. (1996). Int. J. Cancer, 68, 397-405). After loading, the column was washed with 50 ml of equilibration buffer (PBS, 1 mM EDTA, 0.5 M NaCl) . Antibody fragments were then eluted with triethylamine 100 mM, immediately neutralised with 1M Hepes, pH 7, and dialysed against PBS. Affinity measurements by BIAcore were performed with purified antibodies as described (Neri et al. (1997). Nature Biotechnol., 15 1271-1275). Band-shift analysis was performed as described (Neri et al. (1996). Nature Biotechnology, 14, 385-390), using recombinant ED-B fluorescently labeled at the N-terminal extremity (Carnemolla et al. (1996). Int. J. Cancer, 68, 397-405, Neri et al. (1997). Nature Biotechnol., 15 1271-1275) with the infrared fluorophore Cy5 (Amersham). BIAcore analysis does not always allow the accurate determination of kinetic parameters for slow dissociation reactions due to possible rebinding effects, baseline instability and long measurement times needed to ascertain that the dissociation phase follows a single exponential profile. We therefore performed measurements of the kinetic dissociation constant koff by competition experiments (Neri et al. (1996), Trends in Biotechnol., 14, 465-470). In brief, anti-ED-B antibodies (30 nM) were incubated with biotinylated ED-B (10 nM) for 10 minutes, in the presence of M2 anti-FLAG antibody (0.5 μg/ml) and polyclonal anti-mouse IgG (Sigma) which had previously been labeled with a rutenium complex as described (Deaver, D. R. (1995). Nature, 377, 758-760). To this solution, in parallel reactions, unbiotinylated ED-B (1 μM) was added at different times. Streptavidin-coated dynabeads, diluted in Origen Assay Buffer (Deaver, D. R. (1995). Nature, 377, 758-760) were then added (20 μl, 1 mg/ml), and the resulting mixtures analysed with a ORIGEN Analyzer (IGEN Inc. Gaithersburg, Md. USA). This instrument detects an electrochemiluminescent signal (ECL) which correlates with the amount of scFv fragment still bound to the biotinylated ED-B at the end of the competition reaction. Plot of the ECL signal versus competition time yields a profile, that can be fitted with a single exponential with characteristic constant koff.



EXAMPLE 3


Targeting Tumours with a High-affinity Radiolabeled scFv Specific for the ED-B Domain of Fibronectin

[0063] Radioiodinated scFv(L19) or scFv(D1.3) (an irrelevant antibody specific for hen egg lysozyme) were injected intravenously in mice with subcutaneously implanted murine F9 teratocarcinoma, a rapidly growing aggressive tumour. Antibody biodistributions were obtained at different time points (FIG. 1). ScFv(L19) and scFv(D1.3) were affinity purified on an antigen column (Neri et al. (1997, Nature Biotechnol. 15, 1271-1273) and radiolabeled with iodine-125 using the Iodogen method (Pierce, Rockford, Ill., USA). Radiolabeled antibody fragments retained >80% immunoreactivity, as evaluated by loading the radiolabeled antibody onto an antigen column, followed by radioactive counting of the flow-through and eluate fractions. Nude mice (12 weeks old Swiss nudes, males) with subcutaneously-implanted F9 murine teratocarcinoma (Neri et al. (1997) Nature Biotechnol. 15, 1271-1273) were injected with 3 μg (3-4 μCi) of scFv in 100 μl saline solution. Tumour size was 50-250 mg, since larger tumours tend to have a necrotic centre. However, targeting experiments performed with larger tumours (300-600 mg) gave essentially the same results. Three animals were used for each time point. Mice were killed with humane methods, and organs weighed and radioactively counted. Targeting results of representative organs are expressed as percent of the injected dose of antibody per gram of tissue (% ID/g). ScFv(L19) is rapidly eliminated from blood through the kidneys; unlike conventional antibodies, it does not accumulate in the liver or other organs. Eight percent of the injected dose per gram of tissue localises on the tumour already three hours after injection; the subsequent decrease of this value is due to the fact that the tumour doubles in size in 24-48 hours. Tumour:blood ratios at 3, 5 and 24 hours after injection were 1.9, 3.9 and 11.8 respectively for L19, but always below 1.0 for the negative control antibody.


[0064] Radiolabeled scFv(L19) preferentially localises on tumours already few hours after injection, suggesting its usefulness for the immunoscintigraphic detection of angiogenesis in patients.
3TABLE 1Sequences of selected antibody clonesVH chainVL chainAntigenClone       31-33*50-54*      95-98*32*50*91-96*ED-B A2S Y A       A I S G S GGL S IYGN G W Y P W G4S Y A       A I S G S GSF S FYGG G W L P Y E1S Y A       A I S G S GPF P YYGT G R I P P H10S F S       S I R G S SPF P YYCT G R I P P L19S F S       S I R G S SPF P YYYT G R I P PRelevant amino acid positions (*numbering according to ref. 34) of antibody clones isolated from the designed synthetic libraries. Single amino acid codes are used according to standard TUPAC nomenclature.


[0065]

4





TABLE 2










Affinities of anti-ED-B scFv fragments













Clone
kon (s−1M−1)
koff (s−1)B
koff (s−1)C
Kd (M) *







A2
1.5 × 105
2.8 × 10−3

1.9 × 10−8



G4
4.0 × 104
3.5 × 10−3

8.7 × 10−8



E1
1.6 × 105
6.5 × 10−3

4.1 × 10−8



H10
6.7 × 104
5.6 × 10−4
9.9 × 10−5
1.5 × 10−9



L19
1.1 × 105
9.6 × 10−5
6.0 × 10−6
5.4 × 10−11








* Kd = koff / kon. For the high-affinity binders H10 and L19, koff values from BIAcore experiments are not sufficietly reliable due to effects of the negatively-charged carboxylated solid dextran matrix; Kd values are therefore calculated from koff measurements obtained by competition experiments (Experimental Procedures).





koff, kinetic dissociation constant; kon, kinetic association constant; Kd, dissociation constant. B = measured on the BIAcore; C = measured by competition with electrochemiluminescent detection. Values are accurate to +/− 50%, on the basis of the precision of concentration determinations.








[0066]


Claims
  • 1. An antibody with specific affinity for a characteristic epitope of the ED-B domain of fibronectin, wherein the antibody has improved affinity to said ED-B epitope.
  • 2. The antibody according to claim 1, wherein the affinity is in the subnanomolar range.
  • 3. The antibody according to claim l wherein the antibody recognizes ED-B(+) fibronectin.
  • 4. The antibody according to claim 1, wherein said antibody is in the scFv format.
  • 5. The antibody according to claim 4, the antibody being a recombinant antibody.
  • 6. The antibody according to claim 4, wherein the affinity is improved by introduction of a limited number of mutations in its CDR residues.
  • 7. The antibody according to claim 6, wherein the residues are residues 31-33, 50, 52 and 54 of VH and two residues 32 and 50 of its VL domain which have been mutated.
  • 8. The antibody according to claim 1, wherein the antibody binds the ED-B domain of fibronectin with a Kd of 27 to 54 pM, most preferably with a Kd of 54 pM.
  • 9. The antibody according to claim 1, being the antibody L19.
  • 10. The antibody according to claim 1 with the following amino acid sequence:
  • 11. The antibody according to claim 1, wherein the antibody is a functionally equivalent variant form of L19.
  • 12. The antibody according to claim 9, wherein the antibody is radiolabeled.
  • 13. The antibody according to claim 12, wherein the antibody is radioiodinated.
  • 14. Use of an antibody with specific affinity for a characteristic epitope of the ED-B domain of fibronectin, the antibody having improved affinity to said ED-B domain, for rapid targeting markers of angiogenesis.
  • 15. The use as claimed in claim 14 for immunoscintigraphic detection of angiogenesis.
  • 16. The use as claimed in claim 15 for detecting diseases characterized by vascular proliferation such as diabetic retinopathy, age-related macular degeneration or tumours.
  • 17. The use according to claim 14, wherein the antibody localizes the respective tissue three to four hours, most preferably 3 hours after its injection.
  • 18. A diagnostic kit comprising an antibody with specific affinity for a characteristic epitope of the ED-B domain of fibronectin, said antibody having improved affinity to said ED-B domain and one or more reagents necessary for detecting angiogenesis.
  • 19. Use of an antibody with specific affinity for a characteristic epitope of the ED-B domain of fibronectin, said antibody having improved affinity to said ED-B domain for diagnosis and therapy of tumours and diseases characterized by vascular proliferation.
Continuations (1)
Number Date Country
Parent 09075338 May 1998 US
Child 10321558 Dec 2002 US
Continuation in Parts (2)
Number Date Country
Parent 09512082 Feb 2000 US
Child 10321558 Dec 2002 US
Parent 09300425 Apr 1999 US
Child 10321558 Dec 2002 US