WO2005019255A1 - Muteins of tear lipocalin - Google Patents

Muteins of tear lipocalin Download PDF

Info

Publication number
WO2005019255A1
WO2005019255A1 PCT/EP2003/009404 EP0309404W WO2005019255A1 WO 2005019255 A1 WO2005019255 A1 WO 2005019255A1 EP 0309404 W EP0309404 W EP 0309404W WO 2005019255 A1 WO2005019255 A1 WO 2005019255A1
Authority
WO
WIPO (PCT)
Prior art keywords
mutein
sequence
lipocalin
tear lipocalin
protein
Prior art date
Application number
PCT/EP2003/009404
Other languages
French (fr)
Other versions
WO2005019255A8 (en
Inventor
Arne Skerra
Steffen Schlehuber
Original Assignee
Pieris Proteolab Ag
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 Pieris Proteolab Ag filed Critical Pieris Proteolab Ag
Priority to PCT/EP2003/009404 priority Critical patent/WO2005019255A1/en
Priority to AU2003275958A priority patent/AU2003275958A1/en
Priority to SG200701410-3A priority patent/SG131109A1/en
Priority to AU2004266846A priority patent/AU2004266846B2/en
Priority to JP2006524319A priority patent/JP4856543B2/en
Priority to BRPI0413728A priority patent/BRPI0413728B8/en
Priority to CA2536491A priority patent/CA2536491C/en
Priority to DK04764425T priority patent/DK1660528T3/en
Priority to PCT/EP2004/009447 priority patent/WO2005019256A2/en
Priority to AT04764425T priority patent/ATE446313T1/en
Priority to US10/569,134 priority patent/US7585940B2/en
Priority to CN2004800313369A priority patent/CN1871256B/en
Priority to DE602004023725T priority patent/DE602004023725D1/en
Priority to EP04764425A priority patent/EP1660528B1/en
Publication of WO2005019255A1 publication Critical patent/WO2005019255A1/en
Publication of WO2005019255A8 publication Critical patent/WO2005019255A8/en
Priority to HK06106847.6A priority patent/HK1084404A1/en
Priority to US12/461,633 priority patent/US7893208B2/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00

Definitions

  • the present invention relates to novel muteins derived from tear Hpocalin or a functional homologue thereof.
  • the invention relates to a mutein of human tear lipocalin.
  • the invention also refers to a corresponding nucleic acid molecule encoding such a mutein and to a method for its generation.
  • the invention further refers to a method for producing such a mutein.
  • the invention is directed to a pharmaceutical composition comprising such a lipocalin mutein as well as to various use of the mutein.
  • the members of the lipocalin protein family are typically small, secreted proteins which are characterized by a range of different molecular-recognition properties: their ability to bind various, principally hydrophobic molecules (such as retinoids, fatty acids, cholesterols, prostaglandins, biliverdins, pheromones, tastants, and odorants), their binding to specific cell-surface receptors and their formation of macromolecular complexes. Although they have, in the past, been classified primarily as transport proteins, it is now clear that the lipocalins fulfill a variety of physiological functions.
  • lipocalins have also been implicated in the regulation of the immune response and the mediation of cell homoeostasis (reviewed, for example, in Flower, D.R. (1996) Biochem. J. 318, 1-143 and Flower, D.R. et al. (2000) Biochim. Biophys. Acta 1482, 9-24).
  • the lipocalins share unusually low levels of overall sequence conservation, often with sequence identities of less than 20%. In strong contrast, their overall folding pattern is highly conserved.
  • the central part of the lipocalin structure consists of a single eight-stranded anti-parallel ⁇ -sheet closed back on itself to form a continuously hydrogen-bonded ⁇ -barrel.
  • One end of the barrel is sterically blocked by the N-terminal peptide segment that runs across its bottom as well as three peptide loops connecting the ⁇ -strands.
  • the other end of the ⁇ -barrel is open to the solvent and encompasses a target-binding site which is formed by four peptide loops. It is this diversity of the loops in the otherwise rigid lipocalin scaffold that gives rise to a variety of different binding modes each capable of accommodating targets of different size, shape, and chemical character
  • Tear lipocalin Human tear pre-albumin, now called tear lipocalin (TLPC), was originally described as a major protein of human tear fluid (approximately one third of the total protein content) but has recently also identified in several other secretory tissues including prostate, nasal mucosa and tracheal mucosa. Homologous proteins have been found in rat, pig, dog and horse. Tear lipocalin is an unusual lipocalin member because of its high promiscuity for relative insoluble lipids and binding characteristics that differ from other members of this protein family (reviewed in Redl, B. (2000) Biochim. Biophys. Acta 1482, 241-248).
  • lipophilic compounds of different chemical classes such as fatty acids, fatty alcohols, phospholipids, glycolipids and cholesterol are endogenous ligands of this protein.
  • the strength of ligand (target) binding correlates with the length of the hydrocarbon tail both for alkyl amides and fatty acids.
  • tear lipocalin binds most strongly the least soluble lipids (Glasgow, BJ. et al. (1995) Curr. Eye Res. 14, 363-372; Gasymov, O.K. et al. (1999) Biochim. Biophys. Acta 1433, 307-320).
  • tear lipocalin The precise biological function of human tear lipocalin has not been fully elucidated so far and is still a matter of controversy. In tear fluid, it appears to be most important for the integrity of the tear film by removing lipids from the mucous surface of the eye to the liquid phase (reviewed in Gasymov, O.K. et al. (1999), supra). However, it displays additional activities in vitro that are very unusual among lipocalins, namely inhibition of cystein proteinases as well as non-specific endonuclease activity (van't Hof, W. et al. (1997) J. Biol. Chem. 272, 1837-1841; Yusifov, T.N. et al. (2000) Biochem. J. 347, 815-819).
  • tear lipocalin is able to bind several lipid peroxidation products in vitro resulting in the hypothesis that it might function as a physiological oxidative-stress-induced scavenger of potentially harmful lipophilic molecules (Lechner, M. et al. (2001) Biochem. J. 356, 129-135).
  • Proteins which selectively bind to their corresponding targets by way of non-covalent interaction, play a crucial role as reagents in biotechnology, medicine, bioanalytics as well as in the biological and life sciences in general.
  • Antibodies i.e. immunoglobulins
  • immunoglobulins are a prominent example of this class of proteins.
  • immunoglobulins are currently used.
  • the PCT publication WO 99/16873 discloses the class of anticalins®; polypeptides of the lipocalin family, in which amino acid positions in the region of all four peptide loops are mutated, which are arranged at the end of the cylindrical ⁇ - barrel structure encompassing the binding pocket, and which correspond to those segments in the linear polypeptide sequence comprising the amino acid positions 28 to 45, 58 to 69, 86 to 99 and 114 to 129 of the bilin-binding protein of Pieris brassicae.
  • the PCT publication WO 00/75308 discloses muteins of the bilin-binding protein, which specifically bind digoxigenin, whereas the International Patent Applications WO 03/029463 and WO 03/029471 relate to muteins of the human neutrophil gelatinase-associated lipocalin and apolipoprotein D, respectively.
  • specificity as well as folding stability of a lipocalin variant various approaches using different members of the lipocalin family have been proposed (Skerra, A. (2001) Rev. Mol. Biotechnol. 74, 257-275; Schlehuber, S., and Skerra, A. (2002) Biophys. Chem. 96, 213-228) such as the "shaping" of the ligand-binding pocket or the replacement of additional amino acid residues.
  • lipocalin muteins as adapter or linker molecules which may be attached to a given binding partner via binding site I, whereas binding site II is used for screening/selection purposes or the like.
  • fusion proteins comprising two lipocalin muteins of same or different binding specificity which are coupled to each other by a peptide linker.
  • Such fusion proteins also called “duocahns”, are described in WO 99/16873 and also by Schlehuber, S. & Skerra, A. (2001), Biol. Chem. 382, 1335-1342, for example.
  • histamine-binding proteins have been identified in the saliva of Rhipicephalus appendiculatus ticks (Paesen, G.C. et al. (1999) Mol. Cell 3, 661-671). These proteins sequester histamine at the wound site, outcompeting histamine receptors for the ligand in order to suppress inflammation during blood feeding.
  • the crystal structure of these histamine- binding proteins reveals a lipocalin fold novel in containing two binding sites for histamine having different binding affinities.
  • the sites one of which is a typical lipocalin binding site, are orthogonally arranged and highly rigid, forming an unusually polar internal surface that specifically complements the properties of histamine.
  • SHBP A related protein termed SHBP, which is secreted by a rodent- and cattle-feeding tick, binds both histamine and serotonin at the two different binding sites (Sangamnatdej, S. et al. (2002) Insect Mol. Biol. 11, 79-86).
  • the high- affinity binding site lies perpendicular to the long axis of the ⁇ -barrel leading to distortions in the protein structure compared to other lipocalins. Thus, it appears as if such a binding site cannot be engineered in any given lipocalin.
  • the binding sites are rather buried in the core of the ⁇ -barrel there appear to be sterical limitations with regard to ligand size.
  • such a lipocalin mutein is a mutein derived from a polypeptide of tear lipocalin or a functional homologue thereof, wherein the mutein comprises at least two mutated amino acid residues at any sequence position in the N-terminal peptide stretch as well as the three peptide loops BC, DE, and FG arranged at the end of the ⁇ -barrel structure that is located opposite of the natural lipocalin binding pocket, wherein said tear lipocalin or functional homologue thereof has at least 60% sequence homology with human tear lipocalin, and wherein the mutein binds a given target with detectable affinity.
  • this embodiment is based on the finding of the inventors that amino acids in the three loops at the closed end of the internal ligand binding site of a tear lipocalin and/or the N-terminal peptide stretch of the tear lipocalin (cf. Fig. 4) can be mutated in order to obtain lipocalin muteins that bind a given target with determinable affinity.
  • the invention provides a structurally new class of lipocalin muteins with antibody-like binding properties.
  • these muteins can be used in the same way for the generation of new binding proteins with a predetermined specificity as the class of the above mentioned anticalins® (lipocalin muteins which are derived from the proteins of the lipocalin family such as the bilin-binding protein of Pieris brassicae, in which amino acid positions in the four peptide loops positioned on the open end of the ligand binding site are mutated).
  • lipocalin muteins of the present invention are also called anticalins®.
  • a mutein of the invention is also a mutein derived from a polypeptide of tear lipocalin or a functional homologue thereof.
  • the mutein comprises at least two mutated amino acid residues at any sequence position in the four peptide loops AB , CD, EF, and GH encompassing the natural lipocalin binding pocket, wherein said tear lipocalin or functional homologue thereof has at least 60% sequence homology with human tear lipocalin, and wherein the mutein binds a given target with detectable affinity.
  • this embodiment provides for a new class of scaffold in which amino acids in the four loops at the open end of the ligand binding site of the lipocalins can be mutated for the generation of binding molecules against a desired target.
  • the invention relates to a mutein derived from a polypeptide of tear lipocalin or a functional homologue thereof, wherein the mutein comprises at least two mutated amino acid residues at any sequence position in the N-terminal region as well as the three peptide loops BC, DE, and FG arranged at the end of the ⁇ -barrel structure that is located opposite of the natural lipocalin binding pocket, wherein the mutein comprises at least two mutated amino acid residues at any sequence position in the four peptide loops AB, CD, EF, and GH encompassing the natural lipocalin binding pocket, wherein said tear lipocalin or functional homologue thereof has at least 60% sequence homology with human tear lipocalin, and wherein the mutein binds at least one given target with detectable affinity.
  • the invention also provides for the first time a monomeric lipocalin mutein or anticalin® that due to the presence of two binding sites can have binding specifity for two given ligands.
  • a bispecific molecule can be considered to be functionally equivalent to a bispecific antibody molecule such as a bispecific diabody.
  • this new class of bispecific anticalins® has the advantage that it is composed only of one polypeptide chain whereas a diabody consists of two polypeptide chains which are non covalently associated with each other.
  • a bispecific lipocalin mutein of this new class of binding proteins may be used as an adapter molecule.
  • a bispecific lipocalin molecule can cross-link these receptors.
  • An example of such an anticalin® would be a mutein, wherein the first binding site binds to a apoptose receptor such as the CD95 (also known as Fas or Apo 1 receptor) and the second binding site can bind to a cell surface receptor which is expressed on the same cell.
  • Binding of such a bispecific mutein in a bicellular manner may result in mutual cross-linking of the CD95 apoptose receptor and the second cell surface receptor target antigen, which can effectively induce apoptosis of the cells (cf. Jung, G. et al. (2001) Cancer Res. 61, 1846-1848).
  • a bispecific mutein may also have only binding affinity for one given target.
  • Such a mutein may be useful as a molecular storage for drugs that are to be slowly released into the blood stream.
  • mutagenesis means that the experimental conditions are chosen such that the amino acid naturally occurring at a given sequence position of the lipocalin used can be substituted by at least one amino acid that is not present at this specific position in the respective natural polypeptide sequence.
  • mutagenesis also includes the (additional) modification of the length of sequence segments by deletion or insertion of one or more amino acids.
  • one amino acid at a chosen sequence position is replaced by a stretch of three random mutations, leading to an insertion of two amino acid residues compared to the length of (the respective segment) of the wild-type protein.
  • random mutagenesis refers to that no predetermined single amino acid (mutation) is present at a certain sequence position but that at least two amino acids can be incorporated into a selected sequence position during mutagenesis with a certain probability.
  • Such experimental conditions can, for example, be achieved by incorporating codons with a degenerate base composition into a nucleotide acid encoding the respective lipocalin employed.
  • tear lipocalin as used herein is not limited to the human tear lipocalin (SWISS-PROT Data Bank Accession Number M90424) but is meant to include all polypeptides having the structurally conversed lipocalin fold and a sequence homology with respect to the amino acid sequence of the human tear lipocalin of at least 60% .
  • lipocalin fold is used in its regular meaning as used, e.g., in Flower, D.R.
  • the definition of the peptide loops as used in the present invention is also in accordance with the regular meaning of the term lipocalin fold and is as follows and also illustrated in Fig. 4:
  • the peptide loop (segment) AB connects the ⁇ -strands A and B of the cylindrically closed ⁇ -sheet
  • the peptide loop CD connects the ⁇ -strands C and D
  • the peptide loop EF connects the ⁇ -strands E and F
  • the peptide loop GH connects the ⁇ -strands G and H
  • the peptide loop BC connects the ⁇ - strands B and C
  • the loop DE connects the ⁇ -strands D and E
  • the loop FG connects the ⁇ - strands F and G.
  • the loops AB, CD, EF and GH form the known binding site of the lipocalins (which was therefore called the open end), whereas, as found in the present invention, the loops BC, DE and FG can be used together with the N-terminal peptide stretch to form a second binding site which is located at the closed end of the ⁇ -barrel.
  • the term "tear lipocalin” includes structural homologues, already identified or yet to be isolated, from other species which have an amino acid sequence homology of more than about 60 %.
  • the percentage of homology is determined herein using the program BLASTP, version blastp 2.2.5 (November 16, 2002; cf. Altschul, S. F. et al. (1997) Nucleic Acids Res. 25, 3389-3402).
  • the percentage of homology is based on the alignment of the entire polypeptide sequences (cutoff value set to 10 " ) including the propeptide sequences, using the human tear lipocalin as reference in a pairwise comparison. It is calculated as the percentage of numbers of "positives" (homologues amino acids) indicated as result in the BLASTP program output divided by the total number of amino acids selected by the program for the alignment.
  • this total number of selected amino acids can differ from the length of the tear lipocalin (176 amino acids including the propeptide) as it is seen in the following.
  • homologues proteins are Von Ebners gland protein 1 of Rattus norvegicus (VEGP protein; SWISS-PROT Data Bank Accession Numbers P20289) with a sequence homology of ca.
  • Such a structural homologue of the tear lipocalin can be derived from any species, i.e. from prokaryotic as well as from eukaryotic organisms.
  • the structural homologue can be derived from invertebrates as well as vertebrates such as mammals (e.g., human, monkey, dog, rat or mouse) or birds or reptiles.
  • the definition of the mutated sequence positions given for tear lipocalin can be assigned to the other lipocalin with the help of published sequence alignments or alignments methods which are available to the skilled artisan.
  • a sequence alignment can, for example, be carried out as explained in WO 99/16873 (cf. Fig. 3 therein), using an published alignment such as the one in Fig. 1 of Redl, B. (2000) Biochim. Biophys. Acta, 1482, 241-248.
  • the three-dimensional structure of the lipocalins are available structural superpositions can also be used for the determination of those sequence positions that are to be subjected to mutagenesis in the present invention.
  • Other methods of structural analysis such as multidimensional nuclear magnetic resonance spectroscopy can also be employed for this purpose.
  • the homologue of tear lipocalin can also be a mutein protein of tear lipocalin itself, in which amino acid substitutions are introduced at positions other than the positions selected in the present invention.
  • a mutein can be a protein in which positions at the solvent exposed surface of the ⁇ -barrel are mutated compared to the wild type sequence of the tear lipocalin in order to increase the solubility or the stability of the protein.
  • tear lipocalin includes all proteins that have a sequence homology of more than 60%, 70% 80%, 85%, 90% or 95% in relation to the human tear lipocalin (SWISS-PROT Data Bank Accession Number M90424).
  • the mutein as disclosed herein is derived from human tear lipocalin. In other preferred embodiments the mutein is derived from the VEGP protein, VEG protein 2, LCN 1 or ALL 1 protein.
  • the mutein according to the invention typically comprises mutations at any two or more of the sequence positions in the peptide segments corresponding to the sequence positions 7-14, 41-49, 69-77, and 87-98 of the linear polypeptide sequence of human tear lipocalin.
  • the positions 7-14 are part of the N-terminal peptide stretch
  • the positions 41-49 are comprised in the BC loop
  • the positions 60-77 are comprised in the DE loop
  • the positions 87-98 are comprised in the FG loop.
  • the mutations are introduced at those sequence positions which correspond to the positions 8, 9, 10, 11, 12, 13, 43, 45, 47, 70, 72, 74, 75, 90, 92, 94, and 97 of human tear lipocalin.
  • a mutein comprises mutations at 5-10 or 12-16 or all 17 of the sequence positions.
  • a lipocalin mutein comprises mutations at any two or more of the sequence positions in the peptide segments corresponding to the sequence positions 24-36, 53-66, 79-84, and 103-110 of the linear polypeptide sequence of human tear lipocalin.
  • the positions 24-36 are comprised in the AB loop
  • the positions 53-66 are comprised in the CD loop
  • the positions 69-77 are comprised in the EF loop
  • the positions 103-110 are comprised in the GH loop.
  • a mutein having two binding sites comprises mutations at any two or more of the sequence positions in the peptide segments corresponding to the sequence positions 7-14, 41-49, 69-77, and 87-97 of the linear polypeptide sequence of human tear lipocalin and additional mutations at any two or more of the sequence positions in the peptide segments corresponding to the sequence positions 24-36, 53-66, 79-84, and 103-110 of the linear polypeptide sequence of human tear lipocalin.
  • the number of the segments (loops) defined above which are used for mutagenesis can vary (the N-terminal peptide stretch is included in the meaning of the term segment or loop). It is not necessary to mutate all four of these segments altogether of each of two binding sites, for example in a concerted mutagenesis. But it is also possible to introduce mutations only in one, two or three segments of each binding site in order to generate a mutein having detectable affinity to a given target. Therefore, it is possible to subject, for example, only two or three segments at the closed end of the ⁇ -barrel to mutagenesis if a binding molecule with only one engineered binding site is wanted. If this molecule is then wanted to have binding affinity towards a second target, sequence positions in any of the four loops of the second binding site can then be mutated.
  • the lipocalin muteins of the invention may comprise the wild-type (natural) amino acid sequence outside the mutated segments.
  • the lipocalin muteins disclosed herein may also contain amino acid mutations outside the sequence positions subjected to mutagenesis as long as those mutations do not interfere with the binding activity and the folding of the mutein.
  • Such mutations can be accomplished very easily on DNA level using established standard methods (Sambrook, J. et al. (1989) Molecular Cloning: A Laboratory Manual, 2nd Ed., Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY).
  • Possible alterations of the amino acid sequence are insertions or deletions as well as amino acid substitutions. Such substitutions may be conservative, i.e.
  • an amino acid residue is replaced with a chemically similar amino acid residue.
  • conservative substitutions are the replacements among the members of the following groups: 1) alanine, serine, and threonine; 2) aspartic acid and glutamic acid; 3) asparagine and glutamine; 4) arginine and lysine; 5) isoleucine, leucine, methionine, and valine; and 6) phenylalanine, tyrosine, and tryptophan.
  • Such modifications of the amino acid sequence include directed mutagenesis of single amino acid positions in order to simplify sub-cloning of the mutated lipocalin gene or its parts by incorporating cleavage sites for certain restriction enzymes. Furthermore, mutations can be introduced in order to improve certain characteristics of the mutein.
  • the lipocalin muteins of the invention are able to bind the desired target with detectable affinity, i.e. with an affinity constant of preferably at least 10 5 M" 1 . Lower affinities are generally no longer measurable with common methods such as ELIS A and therefore of secondary importance.
  • lipocalin muteins which bind the desired target with an affinity of at least 10 6 M _1 , corresponding to a dissociation constant of the complex of 1 ⁇ M.
  • the binding affinity of a mutein to the desired target can be measured by a multitude of methods such as fluorescence titration, competition ELIS A or surface plasmon resonance.
  • a tear lipocalin mutein of the invention may be used for complex formation with a given target.
  • the target (ligand) may be any chemical compound in free or conjugated form which exhibits features of an immunological hapten, a hormone such as steroid hormones or any biopolymer or fragment thereof, for example, a protein or protein domain, a peptide, an oligodeoxynucleotide, a nucleic acid, an oligo- or polysaccharide or conjugates thereof.
  • the target is a protein.
  • the protein can be any globular soluble protein or a receptor protein, for example, a trans-membrane protein involved in cell signaling, a component of the immune systems such as an MHC molecule or cell surface receptor that is indicative of a specific disease.
  • the mutein may also be able to bind only fragments of a protein.
  • a mutein can bind to a domain of a cell surface receptor, when it is part of the receptor anchored in the cell membrane as well as to the same domain in solution, if this domain can be produced as a soluble protein as well.
  • the invention is by no means limited to muteins that only bind such macromolecular targets. But it is also possible to obtain muteins of tear lipocalin by means of mutagenesis which show specific binding affinity to ligands of low(er) molecular weight such as biotin, fluorescein or digoxigenin.
  • the invention is also directed to lipocalin muteins which are conjugated to a label selected from the group consisting of enzyme labels, radioactive labels, colored labels, fluorescent labels, chromogenic labels, luminescent labels, haptens, digoxigenin, biotin, metal complexes, metals, and colloidal gold.
  • a label selected from the group consisting of enzyme labels, radioactive labels, colored labels, fluorescent labels, chromogenic labels, luminescent labels, haptens, digoxigenin, biotin, metal complexes, metals, and colloidal gold.
  • the mutein may also be conjugated to an organic molecule.
  • organic molecule as used herein preferably denotes an organic molecule comprising at least two carbon atoms, but preferably not more than seven rotatable carbon bonds, having a molecular weight in the range between 100 and 2000 Dalton, preferably 1000 Dalton, and optionally including one or two metal atoms.
  • the lipocalin mutein with any appropriate chemical substance or enzyme, which directly or indirectly generates a detectable compound or signal in a chemical, physical or enzymatic reaction.
  • An example for a physical reaction is the emission of fluorescence upon irradiation or the emission of X-rays when using a radioactive label.
  • Alkaline phosphatase, horseradish peroxidase or ⁇ -galactosidase are examples of enzyme labels which catalyze the formation of chromogenic reaction products.
  • all labels commonly used for antibodies can also be used for conjugation to the muteins of the present invention. Such conjugates can be produced by methods well known in the art.
  • the inventive lipocalin mutein is fused at its N- terminus or its C-terminus to a protein, a protein domain or a peptide such as a signal sequence and/or an affinity tag.
  • the fusion partner may confer new characteristics to the inventive hpocalin mutein such as enzymatic activity or binding affinity for other molecules.
  • suitable fusion proteins are alkaline phosphatase, horseradish peroxidase, gluthation-S-transferase, the albumin-binding domain of protein G, protein A, antibody fragments, oligomerization domains, lipocalin muteins of same or different binding specificity (which results in the formation of "duocahns", cf. Schlehuber, S. & Skerra, A. (2001), Biol. Chem. 382, 1335-1342), or toxins. If two bispecific lipocalin mutein of the inventions (i.e. each of them has two binding sites) are combined into a
  • duocalin a tetravalent molecule is formed. If for example a duocalin is generated from only one mutein having two binding sites that specifically bind biotin, a tetravalent molecule (homodimer) comparable to streptavidin (which is a homotetramer, in which each monomer binds one biotin molecule) can be obtained. Due to expected avidity effects such a mutein might be a useful analytical tools in methods that make use of the detection of biotin groups. Affinity tags such as the Strep-tag ® or Strep-tag ® II (Schmidt, T.G.M. et al. (1996) J. Mol. Biol.
  • the myc-tag, the FLAG-tag, the His 6 -tag or the HA-tag or proteins such as glutathione-S-transf erase also allow easy detection and/or purification of recombinant proteins are further examples of preferred fusion partners.
  • proteins with chromogenic or fluorescent properties such as the green fluorescent protein (GFP) or the yellow fluorescent protein (YFP) are suitable fusion partners for a lipocalin mutein of the invention as well.
  • fusion protein as used herein also comprises lipocalin muteins according to the invention containing a signal sequence.
  • Signal sequences at the N-terminus of a polypeptide direct this polypeptide to a specific cellular compartment, for example the periplasm of E. coli or the endoplasmatic reticulum of eukaryotic cells. A large number of signal sequences is known in the art.
  • a preferred signal sequence for secretion a polypeptide into the periplasm of E. coli is the OmpA-signal sequence.
  • the present invention also relates to nucleic acid molecules (DNA and RNA) comprising nucleotide sequences coding for muteins as described herein. Since the degeneracy of the genetic code permits substitutions of certain codons by other codons specifying the same amino acid, the invention is not limited to a specific nucleic acid molecule encoding a fusion protein of the invention but includes all nucleic acid molecules comprising nucleotide sequences encoding a functional fusion protein.
  • nucleic acid molecule of invention its sequence is derived from the coding sequence of human tear lipocalin.
  • nucleic acid is derived from the VEGP protein, VEG protein 2, LCN 1 or ALL 1 protein
  • the nucleic acid sequence encoding a mutein according to the invention comprises mutations at any two or more of the sequence positions in the peptide segments corresponding to the sequence positions 7-14, 43-49, 70-77, and 87-97 of the linear polypeptide sequence of human tear lipocalin, with the sequence positions corresponding to the positions 8, 9, 10, 11, 12, 13, 43, 45, 47, 70, 72, 74, 75, 90, 92, 94, and 97 ofhuman tear lipocalin being particularly preferred.
  • nucleic acid sequence encoding a mutein according to the invention comprises mutations at any two or more of the sequence positions in the peptide segments corresponding to the sequence positions 24-36, 53-66, 79-84, and 103-110 of the linear polypeptide sequence of human tear lipocalin.
  • nucleic acid molecules encoding a mutein of the invention comprising mutations at any two or more of the sequence positions in the peptide segments corresponding to the sequence positions 7-14, 43-49, 70-77, and 87-97 of the linear polypeptide sequence of human tear lipocalin mutations and additional mutations at any two or more of the sequence positions in the peptide segments corresponding to the sequence positions 24-36, 53-66, 79-84, and 103-110 of the linear polypeptide sequence of human tear lipocalin.
  • the invention as disclosed herein also includes nucleic acid molecules encoding TLPC muteins, which comprise additional mutations outside the segments of experimental mutagenesis. Such mutations are often tolerated or can even prove to be advantageous, for example if they contribute to an improved folding efficiency, protein stability or ligand binding affinity of the mutein.
  • a nucleic acid molecule disclosed in this application may be "operably linked" to a regulatory sequence (or regulatory sequences) to allow expression of this nucleic acid molecule.
  • a nucleic acid molecule such as DNA
  • An operable linkage is a linkage in which the regulatory sequence elements and the sequence to be expressed are connected in a way that enables gene expression.
  • the precise nature of the regulatory regions necessary for gene expression may vary among species, but in general these regions comprise a promoter which, in prokaryotes, contains both the promoter per se, i.e.
  • promoter regions normally include 5 ' non-coding sequences involved in initiation of transcription and translation, such as the -35/-10 boxes and the Shine-Dalgarno element in prokaryotes or the TATA box, CAAT sequences, and 5 -capping elements in eukaryotes. These regions can also include enhancer or repressor elements as well as translated signal and leader sequences for targeting the native polypeptide to a specific compartment of a host cell.
  • the 3' non-coding sequences may contain regulatory elements involved in transcriptional termination, polyadenylation or the like. If, however, these termination sequences are not satisfactory functional in a particular host cell, then they may be substituted with signals functional in that cell.
  • a nucleic acid molecule of the invention can include a regulatory sequence, preferably a promoter sequence.
  • a nucleic acid molecule of the invention comprises a promoter sequence and a transcriptional termination sequence.
  • Suitable prokaryotic promoters are, for example, the tet promoter, the Z ⁇ cUV5 promoter or the T7 promoter. Examples of promoters useful for expression in eukaryotic cells are the SV40 promoter or the CMV promoter.
  • the nucleic acid molecules of the invention can also be comprised in a vector or any other cloning vehicles, such as plasmids, phagemids, phage, baculovirus, cosmids or artificial chromosomes.
  • the nucleic acid molecule is comprised in a phasmid.
  • a phasmid vector denotes a vector encoding the intergenic region of a temperent phage, such as M13 or f 1, or a functional part thereof fused to the cDNA of interest.
  • Such cloning vehicles can include, aside from the regulatory sequences described above and a nucleic acid sequence encoding a lipocalin mutein of the invention, replication and control sequences derived from a species compatible with the host cell that is used for expression as well as selection markers conferring a selectable phenotype on transformed or transfected cells. Large numbers of suitable cloning vectors are known in the art, and are commercially available.
  • the DNA molecule encoding lipocalin muteins of the invention and in particular a cloning vector containing the coding sequence of such a lipocalin mutein can be transformed into a host cell capable of expressing the gene. Transformation can be performed using standard techniques (Sambrook, J. et al. (1989), supra). Thus, the invention is also directed to a host cell containing a nucleic acid molecule as disclosed herein.
  • the transformed host cells are cultured under conditions suitable for expression of the nucleotide sequence encoding a fusion protein of the invention.
  • Suitable host cells can be prokaryotic, such as Escherichia coli (E. coli) or Bacillus subtilis, or eukaryotic, such as Saccharomyces cerevisiae, Pichiapastoris, SF9 or High5 insect cells, immortalized mammalian cell lines (e.g. HeLa cells or CHO cells) or primary mammalian cells.
  • the invention also relates to a method for the generation of a mutein according to the invention or a fusion protein thereof, comprising: (a) subjecting a nucleic acid molecule encoding a tear lipocalin of a functional homologue thereof, wherein said tear lipocalin or functional homologue thereof has at least 60% sequence homology with human tear lipocalin, to mutagenesis at two or more different nucleotide triplets, resulting in one or more mutein nucleic acid molecules(s); (b) expressing the one or more mutein nucleic acid molecule(s) obtained in (a) in a suitable expression system, and (c) enriching at least one mutein having a detectable binding affinity for a given target by means of selection and/or isolation.
  • the nucleic acid molecule can be individually subjected to mutagenesis at two or more different nucleotide triplets in any one, two, three or all four above-mentioned peptide segments arranged at either end of the ⁇ -barrel structure.
  • a mutein or a fusion protein thereof is obtained starting from the nucleic acid encoding tear lipocalin or a functional homologue thereof, which is subjected to mutagenesis and introduced into a suitable bacterial or eukaryotic host organism by means of recombinant DNA technology (as already outlined above).
  • the coding sequence of, for example, human tear lipocalin can serve as a starting point for mutagenesis of the peptide segments selected in the present invention.
  • mutagenesis of the amino acids in the N-terminal peptide stretch and the three peptide loops BC, DE, and FG at the end of the ⁇ -barrel structure that is located opposite of the natural lipocalin binding pocket as well as the four peptide loops AB, CD, EF, and GH encompassing said binding pocket the person skilled in the art has at his disposal the various established standard methods for site-directed mutagenesis (Sambrook, J.
  • a commonly used technique is the introduction of mutations by means of PCR (polymerase chain reaction) using mixtures of synthetic oligonucleotides, which bear a degenerate base composition at the desired sequence positions.
  • PCR polymerase chain reaction
  • nucleotide building blocks with reduced base pair specificity as for example inosine
  • triplet-mutagenesis is another possibility for the introduction of mutations into a chosen sequence segment.
  • This method uses mixtures of different nucleotide triplets each of which codes for one amino acid for the incorporation into the coding sequence.
  • One possible strategy for introducing mutations in the selected regions of the respective polypeptides is based on the use of four oligonucleotides, each of which is partially derived from one of the corresponding sequence segments to be mutated (cf. Fig.3).
  • oligonucleotides each of which is partially derived from one of the corresponding sequence segments to be mutated (cf. Fig.3).
  • a person skilled in the art can employ mixtures of nucleic acid building blocks for the synthesis of those nucleotide triplets which correspond to the amino acid positions to be mutated so that codons encoding all natural amino acids randomly arise, which at last results in the generation of a lipocalin peptide library.
  • the first oligonucleotide corresponds in its sequence - apart from the mutated positions - to the coding strand for the peptide segment to be mutated at the most N-terminal position of the lipocalin polypeptide.
  • the second oligonucleotide corresponds to the non-coding strand for the second sequence segment following in the polypeptide sequence.
  • the third oligonucleotide corresponds in turn to the coding strand for the corresponding third sequence segment.
  • the fourth oligonucleotide corresponds to the non-coding strand for the fourth sequence segment.
  • a polymerase chain reaction can be performed with the respective first and second oligonucleotide and separately, if necessary, with the respective third and fourth oligonucleotide.
  • both of the products can be combined by various known methods into a single nucleic acid comprising the sequence from the first to the fourth sequence segments, in which mutations have been introduced at the selected positions.
  • both of the products can for example be subjected to a new polymerase chain reaction using flanking oligonucleotides as well as one or more mediator nucleic acid molecules, which contribute the sequence between the second and the third sequence segment.
  • This procedure is schematically reproduced in Fig. 3.
  • the person skilled in the art has numerous alternatives at his disposal.
  • the nucleic acid molecules defined above can be connected by ligation with the missing 5 - and 3 -sequences of a nucleic acid encoding a lipocalin polypeptide and/or the vector, and can be cloned in a known host organism.
  • a multitude of established procedures are available for ligation and cloning (Sambrook, J. et al. (1989), supra).
  • recognition sequences for restriction endonucleases also present in the sequence of the cloning vector can be engineered into the sequence of the synthetic oligonucleotides.
  • Longer sequence segments within the gene coding for the protein selected for mutagenesis can also be subjected to random mutagenesis via known methods, for example by use of the polymerase chain reaction under conditions of increased error rate, by chemical mutagenesis or by using bacterial mutator strains. Such methods can also be used for further optimization of the target affinity or specificity of a lipocalin mutein. Mutations possibly occurring outside the segments of experimental mutagenesis are often tolerated or can even prove to be advantageous, for example if they contribute to an improved folding efficiency or folding stability of the lipocalin mutein.
  • the clones carrying the genetic information for the plurality of respective lipocalin muteins, which bind a given target can be selected from the library obtained.
  • Well known techniques can be employed for the selection of these clones, such as phage display (reviewed in Kay, B.K. et al. (1996) supra; Lowman, H. B. (1997) supra or Rodi, D. J. & Makowski, L. (1999) supra), colony screening (reviewed in Pini, A. et al. (2002) Comb. Chem.
  • temperent M13 phage is given as an example of a selection method according to the invention.
  • temperent phage such as f 1 or lytic phage such as T7 may be employed as well.
  • M13 phagemids cf .
  • the mutated lipocalin nucleic acid sequence as a fusion protein with a signal sequence at the N-terminus, preferably the OmpA-signal sequence, and with the capsid protein pHI of the phage M13 or fragments thereof capable of being incorporated into the phage capsid at the C-terminus.
  • the C-terminal fragment ⁇ pIII of the phage capsid protein comprising amino acids 217 to 406 of the wild-type sequence is preferably used to produce the fusion proteins.
  • a C-terminal fragment of pIJJ in which the cysteine residue at position 201 is missing or is replaced by another amino acid.
  • the fusion protein may comprise additional components such as an affinity tag, which allows the immobilization and/or purification of the fusion protein or its parts.
  • a stop codon can be located between the sequence regions encoding the lipocalin or its muteins and the phage capsid gene or fragments thereof, wherein the stop codon, preferably an amber stop codon, is at least partially translated into an amino acid during translation in a suitable suppressor strain.
  • the phagemid vector pTLPC7 (Fig. 1) can be used for the construction of a phage library encoding human tear lipocalin muteins.
  • the inventive nucleic acid molecules coding for the mutated peptide segments are inserted into the vector using the BstXI restriction sites.
  • Recombinant vectors are then transformed into a suitable host strain such as E. coli XLl-Blue.
  • the resulting library is subsequently superinfected in liquid culture with an appropriate M13- helper phage in order to produce functional phage.
  • the recombinant phagemid displays the lipocalin mutein on its surface as a fusion with the coat protein pIJJ or a fragment thereof, while the N-terminal signal sequence of the fusion protein is normally cleaved off. On the other hand, it also bears one or more copies of the native capsid protein pIJJ supplied by the helper phage and is thus capable of infecting a recipient, in general a bacterial strain carrying a F- or F'-plasmid.
  • gene expression of the fusion protein comprised of the lipocalin mutein and the capsid protein pill can be induced, for example by addition of anhydrotetracycline.
  • the induction conditions are chosen such that a substantial fraction of the phage obtained displays at least one lipocalin mutein on their surface.
  • Various methods are known for isolating the phage, such as precipitation with polyethylene glycol. Isolation typically occurs after an incubation period of 6-8 hours.
  • the isolated phage are then subjected to a selection process by incubating them with a given target, wherein the target is present in a form allowing at least a temporary immobilization of those phage displaying muteins with the desired binding activity.
  • a given target wherein the target is present in a form allowing at least a temporary immobilization of those phage displaying muteins with the desired binding activity.
  • the target can be conjugated with a carrier protein such as serum albumin and be bound via this carrier to a protein-binding surface such as polystyrene. Microtiter plates suitable for ELISA techniques or so-called "immunosticks" are preferred.
  • conjugates of the target can also be implemented with other binding groups such as biotin.
  • the target can then be immobilized on surfaces, which will selectively bind this group, such as microtiter plates or paramagnetic particles coated with avidin or streptavidin.
  • the phage particles are captured by binding to the respective target immobilized on the surface. Unbound phage particles are subsequently removed by iterative washing.
  • free target (ligand) molecules can be added to the samples as a competitor.
  • elution can also be achieved by adding proteases or under moderately denaturing conditions, e.g. in the presence of acids, bases, detergents or chaotropic salts.
  • a preferred method is the elution using buffers having pH 2.2, followed by neutralization of the solution.
  • the eluted phage may then be subjected to another selection cycle. Preferably, selection is continued until at least 0.1 % of the clones comprise lipocalin muteins with detectable affinity for the respective target. Depending on the complexity of the library employed 2-8 cycles are required to this end.
  • an E. coli host strain is infected with the phagemids obtained and phagemid DNA is isolated using standard techniques (Sambrook, J. et al. (1989), supra).
  • the mutated sequence fragment or the entire lipocalin mutein nucleic acid sequence can be sub-cloned in any suitable expression vector.
  • the recombinant lipocalin muteins obtained can be purified from their host organism or from a cell lysate by various methods known in the art such as gel filtration or affinity chromatography.
  • lipocalin muteins can also be performed using other methods well known in the art. Furthermore, it is possible to combine different procedures. For example, clones selected or at least enriched by phage display can subsequently be subjected to a colony-screening assay in order to directly isolate a particular lipocalin mutein with detectable binding affinity for a given target. Additionally, instead of generating a single phage library comparable methods can be applied in order to optimize a mutein with respect to its affinity or specificity for the desired target by repeated, optionally limited mutagenesis of its coding nucleic acid sequence.
  • the invention also relates to a method for the production of a mutein of the invention, wherein the mutein, a fragment of the mutein or a fusion protein of the mutein and another polypeptide is produced starting from the nucleic acid coding for the mutein by means of genetic engineering methods.
  • the method can be carried out in vivo, the mutein can for example be produced in a bacterial or eucaryotic host organism and then isolated from this host organism or its culture. It is also possible to produce a protein in vitro, for example by use of an in vitro translation system.
  • a nucleic acid encoding a mutein of the invention is introduced into a suitable bacterial or eukaryotic host organism by means of recombinant DNA technology (as already outlined above).
  • the host cell is first transformed with a cloning vector comprising a nucleic acid molecule encoding a mutein of the invention using established standard methods (Sambrook, J. et al. (1989), supra).
  • the host cell is then cultured under conditions, which allow expression of the heterologous DNA and thus the synthesis of the corresponding polypeptide.
  • the polypeptide is recovered either from the cell or from the cultivation medium.
  • the polypeptide can be preferred to direct the polypeptide to a cell compartment having an oxidizing redox-milieu using an appropriate signal sequence.
  • an oxidizing environment is provided in the periplasm of Gram-negative bacteria such as E. coli or in the lumen of the endoplasmatic reticulum of eukaryotic cells and usually favors the correct formation of the disulfide bonds.
  • the polypeptide can, for instance, be produced in form of inclusion bodies, followed by renaturation in vitro.
  • a further option is the use of specific host strains having an oxidizing intracellular milieu, which thus allow the production of the native protein in the cytosol.
  • a mutein of the invention may not necessarily be generated or produced only by use of genetic engineering. Rather, a lipocalin mutein can also be obtained by chemical synthesis such as Merrifield solid phase polypeptide synthesis. It is for example possible that promising mutations are identified using molecular modeling and then to synthesize the wanted (designed) polypeptide in vitro and investigate the binding activity for a given target.
  • the invention also relates to a pharmaceutical composition
  • a pharmaceutical composition comprising at least one inventive mutein or a fusion protein thereof and a pharmaceutically acceptable excipient.
  • the lipocalin muteins according to the invention can be administered via any parenteral or non- parenteral (enteral) route that is therapeutically effective for proteinaceous drugs.
  • Parenteral application methods comprise, for example, intracutaneous, subcutaneous, intramuscular or intravenous injection and infusion techniques, e.g. in the form of injection solutions, infusion solutions or tinctures, as well as aerosol installation and inhalation, e.g. in the form of aerosol mixtures, sprays or powders.
  • Non-parenteral delivery modes are, for instance, orally, e.g. in the form of pills, tablets, capsules, solutions or suspensions, or rectally, e.g. in the form of suppositories.
  • the muteins of the invention can be administered systemically or topically in formulations containing conventional non-toxic pharmaceutically acceptable excipients or carriers, additives and vehicles as desired.
  • the pharmaceutical is administered parenterally to a mammal, and in particular to humans, with aerosol installation being the most preferable application method due to the low molecular weight of the muteins.
  • the muteins of the present invention can be formulated into compositions using pharmaceutically acceptable ingredients as well as established methods of preparation (Gennaro, A.L. and Gennaro, A.R. (2000) Remington: The Science and Practice of Pharmacy, 20th Ed., Lippincott Williams & Wilkins, Philadelphia, PA).
  • pharmaceutically inert inorganic or organic excipients can be used.
  • pills, powders, gelatin capsules or suppositories for example, lactose, talc, stearic acid and its salts, fats, waxes, solid or liquid polyols, natural and hardened oils.
  • Suitable excipients for the production of solutions, suspensions, emulsions, aerosol mixtures or powders for reconstitution into solutions or aerosol mixtures prior to use include water, alcohols, glycerol, polyols, and suitable mixtures thereof as well as vegetable oils.
  • the pharmaceutical composition may also contain additives, such as, for example, fillers, binders, wetting agents, glidants, stabilizers, preservatives, emulsifiers, and furthermore solvents or solubilizers or agents for achieving a depot effect.
  • additives such as, for example, fillers, binders, wetting agents, glidants, stabilizers, preservatives, emulsifiers, and furthermore solvents or solubilizers or agents for achieving a depot effect.
  • additives such as, for example, fillers, binders, wetting agents, glidants, stabilizers, preservatives, emulsifiers, and furthermore solvents or solubilizers or agents for achieving a depot effect.
  • fusion proteins may be incorporated into slow or sustained release or targeted delivery systems, such as liposomes and microcapsules.
  • the formulations can be sterilized by numerous means, including filtration through a bacteria- retaining filter, or by incorporating sterilizing agents in the form of sterile solid compositions which can be dissolved or dispersed in sterile water or other sterile medium just prior to use.
  • a mutein of the present invention or a fusion protein or a conjugate thereof can be employed in many applications.
  • such a mutein can be used in all applications antibodies are used, except those with specifically rely on the glycosylation of the Fc part.
  • a mutein of the invention can also be used for the targeting of a compound to a preselected site.
  • the mutein is contacted with the compound of interest in order to allow complex formation.
  • the complex comprising the mutein and the compound of interest are delivered the preselected site.
  • This use is in particular suitable, but not restricted to, for delivering a drug (selectively) to the site such an infected body part or organ which is supposed to be treated with the drug.
  • Another use of the inventive muteins is the binding/detection of a given target or target molecule, comprising contacting the mutein with a test sample supposed to contain said target, and detecting of the mutein/target complex by a suitable signal.
  • a mutein can also be used for the separation of a given target, comprising contacting the mutein with a sample supposed to contain said target in order to allow complex formation, and separating the mutein/target complex from the sample.
  • the complex comprising the mutein and the target may be immobilized on any suitable solid phase.
  • the detectable signal can be caused by a label, as explained above, or by a change of physical properties due to the binding, i.e. the complex formation, itself.
  • a label as explained above
  • a change of physical properties due to the binding i.e. the complex formation, itself.
  • plasmon surface resonance the value of which is changed during binding of binding partners from which one is immobilized on a surface such as a gold foil.
  • muteins disclosed herein and its derivatives can thus be used in many fields similar to antibodies or fragments thereof.
  • the muteins can be used for labeling with an enzyme, an antibody, a radioactive substance or any other group having biochemical activity or defined binding characteristics.
  • their respective targets or conjugates or fusion proteins thereof can be detected or brought in contact with them.
  • muteins of the invention can serve to detect chemical structures by means of established analytical methods (e.g. ELISA or Western Blot) or by microscopy or immunosensorics.
  • the detection signal can either be generated directly by use of a suitable mutein conj ugate or fusion protein or indirectly by immunochemical detection of the bound mutein via an antibody.
  • a mutant polypeptide of the invention which binds, for example, tissue- or tumor-specific cellular surface molecules can be generated.
  • Such a mutein may, for example, be employed in conjugated form or as a fusion protein for "tumor imaging" or directly for cancer therapy.
  • target validation i.e. the analysis whether a polypeptide assumed to be involved in the development or progress of a disease or disorder is indeed somehow causative of that disease or disorder.
  • This use for validating a protein as a pharmacological drug target takes advantage of the ability of a mutein of the present invention to specifically recognize a surface area of a protein in its native conformation, i.e. to bind to a native epitope. In this respect, it is to be noted that this ability has been reported only for a limited number of recombinant antibodies.
  • the use of an inventive mutein for validation of a drug target is not limited to the detection of proteins as targets, but also includes the detection of protein domains, peptides, nucleic acid molecules, organic molecules or metal complexes.
  • Figure 1 schematically depicts the phagemid vector pTLPC7
  • Figure 2 schematically depicts the phasmid vector pTLPC6
  • Figure 3 schematically illustrates the generation of the library of tear lipocalin muteins at the nucleic acid level
  • Figure 4 schematically depicts the structure of the lipocalin fold
  • Figure 5 schematically depicts the expression vector pASK75-strepU-CD47
  • Figure 6 shows the polypeptide sequence of mature human tear lipocalin (SWISS-PROT Data Bank Accession Number M90424).
  • Figure 7 shows a schematic drawing of the expression vector pBBP46.
  • Fig.1 shows a schematic drawing of the vector pTLPC7 encoding a fusion protein comprised of the OmpA signal sequence (OmpA), a modified TLPC with the amino acid substitutions Ala5Asp, Ser ⁇ Gly, Asp7Gly, CyslOlSer, and Glul04Gln (for the TLPC cDNA, see Redl et al., supra) and a truncated form of the M13 coat protein pHJ, comprising amino acids 217 to 406 (pin).
  • OmpA OmpA signal sequence
  • a modified TLPC with the amino acid substitutions Ala5Asp, Ser ⁇ Gly, Asp7Gly, CyslOlSer, and Glul04Gln for the TLPC cDNA, see Redl et al., supra
  • a truncated form of the M13 coat protein pHJ comprising amino acids 217 to 406 (pin).
  • the vector further comprises an origin of replication (ori), the intergenic region of the filamentous phage f 1 (fl-IG), the ampicillin resistance gene (bla) coding for ⁇ -lactamase and the tetracycline repressor gene (tetR).
  • An amber stop codon which is partially translated into Gin in SupE amber suppressor host strain, is located between the TLPC coding region and the coding region for the truncated phage coat protein pffl. Both the JSstXI-restriction sites used for the cloning of the mutated gene cassette and the restriction sites flanking the structural gene are labeled.
  • nucleic acid sequence of a Xbal- H d ⁇ i segment of pTLPC7 is shown together with the encoded amino acid sequence in the sequence listing as SEQ ID NO: 1.
  • the vector sequence outside this region is identical with that of pASK75, the complete nucleotide sequence of which is given in the German patent publication DE 44 17 598 Al.
  • Fig. 2 shows a schematic drawing of the vector pTLPC6.
  • pTLPC6 encodes a fusion protein comprised of the OmpA signal sequence, a modified TLPC according to Fig. 1, and the Strep- tag® n affinity tag. Otherwise, the vector is identical to pTLPC7.
  • the nucleic acid sequence of a Xbal- H di ⁇ segment of pTLPC6 is shown together with the encoded amino acid sequence in the sequence listing as SEQ ID NO: 2.
  • the vector sequence outside this region is identical with that of pASK75, the complete nucleotide sequence of which is given in the German patent publication DE 44 17 598 Al.
  • Fig. 3 schematically shows a strategy for the concerted mutagenesis of 17 selected amino acid positions in the modified TLPC by repeated polymerase chain reaction (PCR).
  • PCR polymerase chain reaction
  • a nonsense codon can be substituted by a glutamine-encoding codon, e.g., by site-directed mutagenesis.
  • a nucleic acid fragment with 159 base pairs was amplified (PCR No. 1, A) with the respective primers S ⁇ Q ID NO: 3 and S ⁇ Q ID NO: 4 using the pTLPC6 plasmid-DNA (S ⁇ Q ID NO: 2) as a template.
  • S ⁇ Q ID NO: 2 pTLPC6 plasmid-DNA
  • the ligation efficiency could be improved by purification of the digested PCR-fragment by paramagnetic streptavidin coated beads.
  • the amino acid substitution Glul04Gln as well as the silent mutations in the codon for Ala-3 of the ompA signal sequence, in the codon for Ala21 and Hisl06 were previously accomplished during the construction of pTLPC6 in order to introduce both of the BstXI restriction sites into the TLPC coding sequence.
  • Fig. 4 schematically illustrates the characteristic features of the lipocalin fold (according to Flower, D.R. (1996), supra).
  • the eight ⁇ -strands of the antiparallel ⁇ -sheet which form the ⁇ - barrel) are shown as arrows and labeled A to H (a ninth ⁇ -strand, designated I which is additionally present in some lipocalins, is also schematically shown).
  • the hydrogen-bonded connection of two strands is indicated by a pair of dotted lines between them.
  • the connecting loops are shown as solid curved lines.
  • the two ends of the ⁇ -barrel are topologically distinct.
  • One end has four ⁇ -hairpins (loops AB, CD, EF and GH), the opening of the known ligand binding site of the lipocalins is here and called the open end.
  • the other end of the ⁇ -barrel has three loops (BC, DE and FG) which together with the N-terminal polypeptide region build the closed end and are used in the present invention to introduce an alternative binding site.
  • the parts which form the three main structurally conserved regions (SCRs) of the fold, SCRl , SCR2 and SCR3, are marked as boxes.
  • Fig. 5 shows a schematic drawing of the expression vector pASK75-strepn-CD47.
  • pASK75- strep ⁇ CD47 codes for a fusion protein made of the OmpA signal sequence, a modified extracellular domain of human CD47 comprising amino acids 1 to 119 of the mature protein with the amino acid substitution Cys 15 to Ala and the Strep-tag® ⁇ affinity tag. All further genetic elements are identical with the generic vector pASK75.
  • a relevant segment from the nucleic acid sequence of pASK75-strepH-CD47 is reproduced together with the encoded amino acid sequence in the sequence listing as SEQ ID NO: 10. The segment begins with the Xbal restriction site and ends with the H dUI restriction site.
  • the vector elements outside this region are identical with the vector pASK75, the complete nucleotide sequence of which is exhibited in the German patent publication DE 44 17 598 Al.
  • Fig. 6 shows the polypeptide sequence of mature human tear lipocalin (SWISS-PROT Data Bank Accession Number M90424, 158 amino acids, cf. also Redl B. (2000) Biochim. Biophys. Acta, supra).
  • a human protein that was modified as follows was used in the following examples for the generation of lipocalin muteins.
  • Second, the last two C-terminal amino acid residues (SD) were also deleted.
  • the wild type sequence at sequence positions 5 to 7 (ASD) was changed to GGD.
  • Fig.7 shows a schematic drawing of the expression vector pBBP46.
  • pBBP46 codes for a fusion protein of the OmpA signal sequence and the T7 detection tag (T7) with a modified bilin binding protein of Pieris brassicae (cf. SEQ ID NO: 11) including the C-terminal Strep-tag® ⁇ .
  • This structural gene is followed by the dsbC structural gene (including its ribosomal binding site) from E. coli (Zapun et al., Biochemistry 34 (1995), 5075-5089) as a second cistron (dsbC).
  • dsbC a second cistron
  • the segment begins with the Xbal restriction site and ends with the Hz ⁇ dlTl restriction site.
  • the vector elements outside this region are identical with the vector pASK75, the complete nucleotide sequence of which is exhibited in the German patent publication DE 44 17 598 Al.
  • Example 1 Generation of a library with about 10 billion independent TLPC muteins
  • the reaction mixture contained 10 ⁇ l 10 x Taq buffer (100 mM Tris/HCl pH 9.0, 500 mM KC1, 15 mM MgCl 2 , 1% v/v Triton X-100) and 2 ⁇ l dNTP-Mix (10 mM dATP, dCTP, dGTP, dTTP).
  • a 2000 ⁇ l mixture was prepared, wherein approximately 1000 fmol of both of these respective fragments were used as templates, in the presence of 1000 pmol of each of the assembly primers SEQ ID NO: 7, SEQ ID NO: 8 and 20 pmol of the mediating primer SEQ ID NO: 9.
  • Both assembly primers had a biotin group at their 5'-ends allowing the purification of the PCR-product after Z?,stXI cleavage via streptavidin-coated paramagnetic beads.
  • this fragment representing the library of TPLC muteins in nucleic acid form was first cut with the restriction enzyme .BstXI (Promega) according to the instructions of the manufacturer and then purified by preparative agarose gel electrophoresis as described above, resulting in a double stranded DNA-fragment of 303 nucleotides in size.
  • DNA-fragments not or incompletely digested were removed via their 5 '-biotin tags using streptavidin-coated paramagnetic beads (Merck). Therefore, 200 ⁇ l of the commercially available suspension of the paramagnetic particles in a concentration of 10 mg/ml were washed three times with 100 ⁇ l TE-buffer.
  • the particles were then drained and mixed with 100 pmol of the DNA-fragment in 100 ⁇ l TE-buffer for 15 minutes at room temperature.
  • the paramagnetic particles were collected at the wall of the Eppendorf vessel with the aid of a magnet and the supernatant containing the purified DNA fragment was recovered for further use in the following ligation reaction.
  • the DNA of the vector pTLPC7 (Fig.2) was cut with BstXI as described above and the larger of the two resulting fragments (3907 bp) was purified by preparative agarose gel electrophoresis.
  • DNA was dissolved to a final concentration of 200 ⁇ g/ml in a total volume of 416.5 ⁇ l water.
  • the culture was cooled for 30 minutes on ice and subsequently centrifuged for 15 minutes at 4000 g and 4°C.
  • the cells were washed twice with 500 ml ice-cold 10% w/v glycerol and finally re- suspended in 2 ml of ice-cold GYT-medium (10% w/v glycerol, 0.125% w/v yeast extract, 0.25% w/v tryptone).
  • the cells were then aliquoted (200 ⁇ l), shock-frozen in liquid nitrogen and stored at -80°C.
  • the Micro Pulser system (BioRad) was used in conjunction with cuvettes from the same vendor (electrode separation 2 mm) for electroporation. All steps were carried out at room temperature employing pre-chilled cuvettes at a temperature of -20°C. Each 10 ⁇ l of the DNA solution (2 ⁇ g) was mixed with 100 ⁇ l of the cell suspension, incubated for 1 minute on ice, and transferred to the pre-chilled cuvette. Electroporation was performed (5 ms, 12.5 kV/cm) and the suspension was immediately diluted in 2 ml SOC-medium, followed by incubation for 60 minutes at 37°C and 140 rpm.
  • the culture was diluted in 4 1 2 x YT-medium containing 100 ⁇ g/ml ampicillin (2 YT/Amp) resulting in an OD 55 o of 0.26.
  • 2 YT/Amp 100 ⁇ g/ml ampicillin
  • Example 2 Selection and identification of TLPC muteins with determinable binding affinity towards a given target
  • muteins of the invention with determinable binding affinity towards a given target can be carried in accordance with any protocol known to the skilled persons, for example, in accordance the respective protocols described in any of the PCT publications WO 99/16873, WO 00/75308, WO 03/029463 and WO 03/029471 as well the protocols described above.
  • TLPC muteins can be presented and selected by means of phagemids to a given target that is immobilized on a solid phase. Then, selected muteins can be identified.
  • E. coli JM83 (Yanisch-Perron et al. (1985) Gene 33, 103-119) was transformed with the expression plasmid pASK75-strepII-CD47 (Fig. 5) harboring the cDNA encoding the extracellular domain of CD47 (AS 1-119 of the mature protein) with a single mutation resulting in a Cys ⁇ Ala exchange at amino acid position 15 (for the CD47 cDNA, see Lindberg et al. (1993) J. Cell Biol. 123, 485-496).
  • LB/Amp 100 ml LB-medium containing 100 ⁇ g/ml ampicillin
  • LB/Amp 100 ml LB-medium containing 100 ⁇ g/ml ampicillin
  • the cells from one flask were centrifuged (10 minutes, 5500 g, 4°C) and re-suspended in 20 ml periplasmic release buffer (100 mM Tris/HCl pH 8.0, 500 mM sucrose, 1 mMEDTA), followed by cooling on ice for 30 minutes.
  • the spheroplasts were removed in two successive centrifugation steps (25 minutes, 5300 g, 4°C and 10 minutes, 27000 g, 4°C, respectively).
  • the supernatant containing the periplasmic protein extract was dialyzed against SA-buffer (100 mM Tris/HCl pH 8.0, 150 mM NaCl, 1 mM EDTA), sterile-filtered, and subjected to chromatographic purification.
  • the purification was performed by using the Strep-Tag® ⁇ -affinity tag (Skerra and Schmidt (2000) Methods Enzymol. 326, 271-304) located at the C-terminus of the CD47 protein and by employing Streptactin Superflow material (IB A).
  • a chromatographic column with abed volume of 8 ml was filled with this affinity matrix and equilibrated with 20 ml SA-buffer at 4°C at a flow rate of 60 ml/h. Chromatography was monitored by measuring the absorption at 280 nm of the eluate in a flow-through photometer.
  • the column was washed with SA-buffer until reaching the base line and the bound CD47 was subsequently eluted with ca. 15 ml of a solution of 2.5 mM D-desthiobiotin (IB A) in SA-buffer collecting fractions of the eluate.
  • the fractions containing purified CD47 were analyzed via SDS- polyacrylamide gel electrophoresis (Fling und Gregerson (1986) Anal. Biochem. 155, 83-88), combined and dialyzed against an appropriate buffer, if needed.
  • the protein yield was approximately 370 ⁇ g per 1 1 culture.
  • Example 2.2 Phagemid presentation and selection of TPLC muteins against the extracellular domain of human CD47 employing polystyrol sticks
  • a 2 ml aliquot of the stored phagemids from Example 1 containing a suitable number of phagemids can be centrifuged (30 minutes, 21460 g, 4 °C), the supernatant can then be removed, and the sedimented phagemid particles then dissolved in a buffer such as 1 ml PBS (4 mM KH 2 PO , 16 mM Na 2 HPO , 115 mM NaCl, pH 7.4). After incubation for 30 minutes on ice the solution is centrifuged (5 minutes, 18500 g, 4 °C) to remove residual aggregates.
  • a buffer such as 1 ml PBS (4 mM KH 2 PO , 16 mM Na 2 HPO , 115 mM NaCl, pH 7.4
  • Immuno-sticks can be used for the affinity enrichment of the recombinant phagemids carrying the BBP mutein fusion proteins. These sticks are first coated for a suitable period of time (for instance) overnight at 4 °C with 800 ⁇ l of the extracellular domain of human CD47 (CD47) (100 ⁇ g/ml) in SA-buffer as a target protein. Unoccupied binding sites on the surface of the Immuno-Stick are then saturated by incubation with 1.2 ml 2 % w/v BSA in PBST (PBS with 0.1 % v/v Tween 20) for 2 hours at room temperature.
  • PBST PBS with 0.1 % v/v Tween 20
  • the Immuno-stick can be briefly washed for example three or four times with 1.2 ml PBST and subsequently incubated with a mixture of 500 ⁇ l of the phagemid solution (ca. 5»10 12 cfu) and of 250 ⁇ l of PBS containing 6 % w/v BSA and 0.3 % v/v Tween 20 for 1 hour at room temperature.
  • phagemids For the removal of unbound phagemids, repeated washing can be performed, for examples six to eight times, each time with 950 ⁇ l PBST for 2 minutes. Adsorbed phagemids are then finally eluted by treating the Immuno-stick for 10 minutes with 950 ⁇ l 0.1 M glycine/HCl pH 2.2, followed by immediate neutralisation of the pH of the elution fraction by mixing it with 150 ⁇ l 0.5 M Tris.
  • the cells infected with the phagemids are subsequently sedimented (2 minutes, 4420 g, 4 °C), resuspended in 600 ⁇ l of the culture medium, and plated out onto three agar plates with LB/Amp-medium (LB/Amp agar; 145 mm diameter).
  • the cells are scraped from the agar plates, each with addition of 10 ml 2xYT/Amp, are transferred to a sterile Erlenmeyer-flask, and are shaken for 30 minutes at 37 °C, 140 rpm for complete suspension.
  • 50 ml of 2xYT/Amp can be inoculated to an OD 55 0 of ca. 0.08 with an appropriate volume of this suspension and incubated at 37 °C, 160 rpm until OD550 reached 0.5.
  • VCS-M13 helper phage (Stratagene) at a moi of approximately 10
  • the culture can be shaken for additional 45 minutes at 37 °C, 140 rpm.
  • Kanamycin 70 ⁇ g/ml
  • the incubator temperature lowered to 26 °C and, after 10 minutes, anhydrotetracycline is added at 25 ⁇ g/1 (6.2 ⁇ l of a 0.2 mg/ml stock solution in DMF) in order to induce gene expression. Incubation can continue for another 12-15 hours at 26 °C, 140 rpm.
  • the cells are sedimented by centrifugation (15 minutes, 12100 g, 4 °C) and the supernatant containing the phagemid particles sterile-filtered (0.45 ⁇ m), mixed with 1/4 volume (12.5 ml) 20 % w/v PEG 8000, 15 % w/v NaCl, and incubated on ice for 1 hour. After centrifugation (20 minutes, 18000 g, 4 °C) the precipitated phagemid particles are dissolved in 2 ml of cold BBS/E. The solution can be distributed to two 1.5 ml reaction vessels and incubated on ice for 30 minutes. After centrifugation of undissolved components (5 minutes, 21460 g, 4 °C) each supernatant is transferred to a new reaction vessel.
  • Example 2.3 Identification of CD47-binding BBP muteins by use of a high- throughput ELISA screening method
  • a vector analogues to the vector the pBBP46 (Fig.7) can be used.
  • the gene cassette containing the TLPC between the two BstXI cleavage sites can be subcloned from the vector pTLPC7 into the vector analogues to pBBP46 (Fig. 7).
  • the plasmid DNA is isolated from the mixture of the E. coli clones obtained by infection with the phagemids from Example 2.2 eluted as a result of the last selection cycle, using the Plasmid Miniprep Spin kit (Genomed).
  • the DNA is cut with the restriction enzyme BstXI and the smaller of the two fragments is purified by preparative agarose-gel electrophoresis as described in Example 1.
  • the DNA of the vector that is analogues to pBBP46 is likewise cut with BstXI and the larger of the two fragments (4165 bp) was isolated in the same way.
  • each 50 fmol of the two DNA-fragments are mixed with 3 Weiss Units T4 DNA ligase (Promega) in a total volume of 20 ⁇ l (30 mM Tris/HCl pH 7.8, 10 mM MgCl 2 , 10 mM DTT, 1 mM ATP), followed by incubation for 2 h at 22 °C.
  • E. coli TG1-F E. coli K12 TGI, which had lost its episome
  • E. coli K12 TGI which had lost its episome
  • Single E. coli colonies obtained after the transformation harbouring the respective TLPC plasmids coding for the TLPC muteins are picked from these agar plates into 70 ⁇ l per well 2xYT/Amp in flat bottom 384 well plates (Greiner) by means of an automated colony picker (Genetix) and grown overnight at 37 °C at 700 rpm on a benchtop shaker (Bilhler) in a humidified incubator (MMM Medcenter) at 60 % relative humidity (rH).
  • the cultures can be diluted 1:100 into 100 ⁇ l 2xYT/Amp in round bottom 96 well plates (Nunc) by means of a 96 pin replicating head (Genetix) and grown for ca.
  • Recombinant TLPC muteins are produced in the 96 well plates by adding 20 ⁇ l per well of 1.2 ⁇ g/ml anhydrotetracyclin in 2xYT (obtained by diluting a 2 mg/ml stock solution 1:1667 in 2xYT; final concentration 0.2 ⁇ g/ml) to the bacterial cultures and incubation overnight at 22 °C and 700 rpm at 60 % rH.
  • lysis buffer 400 mM Na-borate pH 8.0, 320 mM NaCl, 4 mM EDTA, 0.3 % w/v lysozyme
  • 40 ⁇ l of lysis buffer 400 mM Na-borate pH 8.0, 320 mM NaCl, 4 mM EDTA, 0.3 % w/v lysozyme
  • 40 ⁇ l/well PBS containing 10 % w/v BSA and 0.05 % v/v Tween 20 (final concentration 2 % BSA) for 1 h at 22 °C and 700 rpm at 60 % rH.
  • the crude cell extracts containing the TLPC muteins are tested for their reactivity with the prescribed target protein CD47 and the unrelated control protein aldolase
  • the wells can be incubated with 10 ⁇ l of the cell extract from above for 1 hour at room temperature. Subsequently, plates are washed again five times and 10 ⁇ l of an anti-T7 monoclonal antibody- HRP-conjugate (Amersham), diluted 1 :5000 in PBST/0.05 containing 0.5 % w/v non-fat dry milk powder (Vitalia), is added to each well and incubated for 1 hour at room temperature.
  • an anti-T7 monoclonal antibody- HRP-conjugate Amersham
  • diluted 1 :5000 in PBST/0.05 containing 0.5 % w/v non-fat dry milk powder (Vitalia)
  • TLPC muteins that showed a binding signal on the prescribed target protein (CD47) which exceeded the binding signal on the unrelated control protein (aldolase) at least by a factor of three (signal-to-control ratio of at least 3) can be subsequently subjected to a secondary high-throughput ELISA screening experiment in order to study the binding specificity of these muteins in greater detail. Therefore, these clones are transferred from the 384 well master plates described above onto LB/Amp agar, and grown overnight at 37 °C.
  • This conjugate is prepared by reacting RNaseA at a twofold molar ratio of digoxigenin-3-O- methylcarbonyl- ⁇ -amidocaproic acid-N-hydroxy-succinimide ester (DIG-NHS; Roche) according to the instructions of the manufacturer. Excess reactant is removed from the RNaseA-conjugate by means of size exclusion chromatography using a HiTrap column (Amersham) according to the instructions of the manufacturer employing PBS as running buffer.
  • DIG-NHS digoxigenin-3-O- methylcarbonyl- ⁇ -amidocaproic acid-N-hydroxy-succinimide ester
  • the nucleotide sequence of chosen TLPC clones can then be determined. Furthermore, these clones can then be chosen for the determination of their binding affinities for CD47.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Zoology (AREA)
  • Biochemistry (AREA)
  • Genetics & Genomics (AREA)
  • Toxicology (AREA)
  • Biophysics (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Molecular Biology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Oncology (AREA)
  • Communicable Diseases (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
  • Investigating Or Analysing Biological Materials (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)

Abstract

The present invention relates to novel muteins derived from tear lipocalin or a functional homologue thereof. In particular, the invention relates to a mutein of human tear lipocalin. The invention also refers to a corresponding nucleic acid molecule encoding such a mutein and to a method for its generation. The invention further refers to a method for producing such a mutein. Finally, the invention is directed to pharmaceutical composition comprising such a lipocalin mutein as well as to various use of the mutein.

Description

MUTEINS OF TEAR LIPOCALIN
The present invention relates to novel muteins derived from tear Hpocalin or a functional homologue thereof. In particular, the invention relates to a mutein of human tear lipocalin. The invention also refers to a corresponding nucleic acid molecule encoding such a mutein and to a method for its generation. The invention further refers to a method for producing such a mutein. Finally, the invention is directed to a pharmaceutical composition comprising such a lipocalin mutein as well as to various use of the mutein.
The members of the lipocalin protein family (Pervaiz, S., and Brew, . (1987) FASEB J. 1, 209- 214) are typically small, secreted proteins which are characterized by a range of different molecular-recognition properties: their ability to bind various, principally hydrophobic molecules (such as retinoids, fatty acids, cholesterols, prostaglandins, biliverdins, pheromones, tastants, and odorants), their binding to specific cell-surface receptors and their formation of macromolecular complexes. Although they have, in the past, been classified primarily as transport proteins, it is now clear that the lipocalins fulfill a variety of physiological functions. These include roles in retinol transport, olfaction, pheromone signaling, and the synthesis of prostaglandins. The lipocalins have also been implicated in the regulation of the immune response and the mediation of cell homoeostasis (reviewed, for example, in Flower, D.R. (1996) Biochem. J. 318, 1-143 and Flower, D.R. et al. (2000) Biochim. Biophys. Acta 1482, 9-24).
The lipocalins share unusually low levels of overall sequence conservation, often with sequence identities of less than 20%. In strong contrast, their overall folding pattern is highly conserved. The central part of the lipocalin structure consists of a single eight-stranded anti-parallel β-sheet closed back on itself to form a continuously hydrogen-bonded β-barrel. One end of the barrel is sterically blocked by the N-terminal peptide segment that runs across its bottom as well as three peptide loops connecting the β-strands. The other end of the β-barrel is open to the solvent and encompasses a target-binding site which is formed by four peptide loops. It is this diversity of the loops in the otherwise rigid lipocalin scaffold that gives rise to a variety of different binding modes each capable of accommodating targets of different size, shape, and chemical character
(reviewed, e.g., in Flower, D.R. (1996), supra; Flower, D.R. et al. (2000), supra, or Skerra, A. (2000) Biochim. Biophys. Acta 1482, 337-350).
Human tear pre-albumin, now called tear lipocalin (TLPC), was originally described as a major protein of human tear fluid (approximately one third of the total protein content) but has recently also identified in several other secretory tissues including prostate, nasal mucosa and tracheal mucosa. Homologous proteins have been found in rat, pig, dog and horse. Tear lipocalin is an unusual lipocalin member because of its high promiscuity for relative insoluble lipids and binding characteristics that differ from other members of this protein family (reviewed in Redl, B. (2000) Biochim. Biophys. Acta 1482, 241-248). A remarkable number of lipophilic compounds of different chemical classes such as fatty acids, fatty alcohols, phospholipids, glycolipids and cholesterol are endogenous ligands of this protein. Interestingly, in contrast to other lipocalins the strength of ligand (target) binding correlates with the length of the hydrocarbon tail both for alkyl amides and fatty acids. Thus, tear lipocalin binds most strongly the least soluble lipids (Glasgow, BJ. et al. (1995) Curr. Eye Res. 14, 363-372; Gasymov, O.K. et al. (1999) Biochim. Biophys. Acta 1433, 307-320).
The precise biological function of human tear lipocalin has not been fully elucidated so far and is still a matter of controversy. In tear fluid, it appears to be most important for the integrity of the tear film by removing lipids from the mucous surface of the eye to the liquid phase (reviewed in Gasymov, O.K. et al. (1999), supra). However, it displays additional activities in vitro that are very unusual among lipocalins, namely inhibition of cystein proteinases as well as non-specific endonuclease activity (van't Hof, W. et al. (1997) J. Biol. Chem. 272, 1837-1841; Yusifov, T.N. et al. (2000) Biochem. J. 347, 815-819). Recently, it has been demonstrated that tear lipocalin is able to bind several lipid peroxidation products in vitro resulting in the hypothesis that it might function as a physiological oxidative-stress-induced scavenger of potentially harmful lipophilic molecules (Lechner, M. et al. (2001) Biochem. J. 356, 129-135).
Proteins, which selectively bind to their corresponding targets by way of non-covalent interaction, play a crucial role as reagents in biotechnology, medicine, bioanalytics as well as in the biological and life sciences in general. Antibodies, i.e. immunoglobulins, are a prominent example of this class of proteins. Despite the manifold needs for such proteins in conjunction with recognition, binding and/or separation of ligands/targets, almost exclusively immunoglobulins are currently used. The application of other proteins with defined ligand-binding characteristics, for example the lectins, has remained restricted to special cases.
Rather recently, members of the lipocalin family have become subject of research concerning proteins having defined ligand-binding properties. The PCT publication WO 99/16873 discloses the class of anticalins®; polypeptides of the lipocalin family, in which amino acid positions in the region of all four peptide loops are mutated, which are arranged at the end of the cylindrical β- barrel structure encompassing the binding pocket, and which correspond to those segments in the linear polypeptide sequence comprising the amino acid positions 28 to 45, 58 to 69, 86 to 99 and 114 to 129 of the bilin-binding protein of Pieris brassicae. The PCT publication WO 00/75308 discloses muteins of the bilin-binding protein, which specifically bind digoxigenin, whereas the International Patent Applications WO 03/029463 and WO 03/029471 relate to muteins of the human neutrophil gelatinase-associated lipocalin and apolipoprotein D, respectively. In order to further improve and fine tune ligand affinity, specificity as well as folding stability of a lipocalin variant various approaches using different members of the lipocalin family have been proposed (Skerra, A. (2001) Rev. Mol. Biotechnol. 74, 257-275; Schlehuber, S., and Skerra, A. (2002) Biophys. Chem. 96, 213-228) such as the "shaping" of the ligand-binding pocket or the replacement of additional amino acid residues.
However, for various applications it could also be advantageous to have more than one binding site per molecule available - either the natural binding pocket plus an engineered additional (protein)-binding site or two different engineered binding sites. For example, it could be considered to use lipocalin muteins as adapter or linker molecules which may be attached to a given binding partner via binding site I, whereas binding site II is used for screening/selection purposes or the like. One possibility to achieve this goal, is the use of fusion proteins comprising two lipocalin muteins of same or different binding specificity which are coupled to each other by a peptide linker. Such fusion proteins, also called "duocahns", are described in WO 99/16873 and also by Schlehuber, S. & Skerra, A. (2001), Biol. Chem. 382, 1335-1342, for example.
Recently high-affinity histamine-binding proteins have been identified in the saliva of Rhipicephalus appendiculatus ticks (Paesen, G.C. et al. (1999) Mol. Cell 3, 661-671). These proteins sequester histamine at the wound site, outcompeting histamine receptors for the ligand in order to suppress inflammation during blood feeding. The crystal structure of these histamine- binding proteins reveals a lipocalin fold novel in containing two binding sites for histamine having different binding affinities. The sites, one of which is a typical lipocalin binding site, are orthogonally arranged and highly rigid, forming an unusually polar internal surface that specifically complements the properties of histamine. A related protein termed SHBP, which is secreted by a rodent- and cattle-feeding tick, binds both histamine and serotonin at the two different binding sites (Sangamnatdej, S. et al. (2002) Insect Mol. Biol. 11, 79-86). The high- affinity binding site lies perpendicular to the long axis of the β-barrel leading to distortions in the protein structure compared to other lipocalins. Thus, it appears as if such a binding site cannot be engineered in any given lipocalin. On the other hand, since the binding sites are rather buried in the core of the β-barrel there appear to be sterical limitations with regard to ligand size.
Thus, there remains a need for the generation of binding proteins that uses different binding sites and/or alternative lipocalin scaffolds, simply for the reason to have more options for practical realisation.
Accordingly, it is an object of the invention to provide alternative lipocalin muteins having binding affinity to a given target.
This object is accomplished by a lipocalin mutein having the features of the independent claims as well as the method for its generation.
In one embodiment such a lipocalin mutein is a mutein derived from a polypeptide of tear lipocalin or a functional homologue thereof, wherein the mutein comprises at least two mutated amino acid residues at any sequence position in the N-terminal peptide stretch as well as the three peptide loops BC, DE, and FG arranged at the end of the β-barrel structure that is located opposite of the natural lipocalin binding pocket, wherein said tear lipocalin or functional homologue thereof has at least 60% sequence homology with human tear lipocalin, and wherein the mutein binds a given target with detectable affinity.
In more illustrative terms, this embodiment is based on the finding of the inventors that amino acids in the three loops at the closed end of the internal ligand binding site of a tear lipocalin and/or the N-terminal peptide stretch of the tear lipocalin (cf. Fig. 4) can be mutated in order to obtain lipocalin muteins that bind a given target with determinable affinity. Thus, the invention provides a structurally new class of lipocalin muteins with antibody-like binding properties. This means that these muteins can be used in the same way for the generation of new binding proteins with a predetermined specificity as the class of the above mentioned anticalins® (lipocalin muteins which are derived from the proteins of the lipocalin family such as the bilin-binding protein of Pieris brassicae, in which amino acid positions in the four peptide loops positioned on the open end of the ligand binding site are mutated). For this reason, these new lipocalin muteins of the present invention are also called anticalins®.
In another embodiment, a mutein of the invention is also a mutein derived from a polypeptide of tear lipocalin or a functional homologue thereof. In this case, the mutein comprises at least two mutated amino acid residues at any sequence position in the four peptide loops AB , CD, EF, and GH encompassing the natural lipocalin binding pocket, wherein said tear lipocalin or functional homologue thereof has at least 60% sequence homology with human tear lipocalin, and wherein the mutein binds a given target with detectable affinity. Accordingly, this embodiment provides for a new class of scaffold in which amino acids in the four loops at the open end of the ligand binding site of the lipocalins can be mutated for the generation of binding molecules against a desired target.
In yet another embodiment the invention relates to a mutein derived from a polypeptide of tear lipocalin or a functional homologue thereof, wherein the mutein comprises at least two mutated amino acid residues at any sequence position in the N-terminal region as well as the three peptide loops BC, DE, and FG arranged at the end of the β-barrel structure that is located opposite of the natural lipocalin binding pocket, wherein the mutein comprises at least two mutated amino acid residues at any sequence position in the four peptide loops AB, CD, EF, and GH encompassing the natural lipocalin binding pocket, wherein said tear lipocalin or functional homologue thereof has at least 60% sequence homology with human tear lipocalin, and wherein the mutein binds at least one given target with detectable affinity.
Thus, the invention also provides for the first time a monomeric lipocalin mutein or anticalin® that due to the presence of two binding sites can have binding specifity for two given ligands. Such a bispecific molecule can be considered to be functionally equivalent to a bispecific antibody molecule such as a bispecific diabody. However, compared to a bispecific diabody (or antibody fragment in general), this new class of bispecific anticalins® has the advantage that it is composed only of one polypeptide chain whereas a diabody consists of two polypeptide chains which are non covalently associated with each other.
A bispecific lipocalin mutein of this new class of binding proteins may be used as an adapter molecule. For example, when having binding affinity to two different receptors, such a bispecific lipocalin molecule can cross-link these receptors. An example of such an anticalin® would be a mutein, wherein the first binding site binds to a apoptose receptor such as the CD95 (also known as Fas or Apo 1 receptor) and the second binding site can bind to a cell surface receptor which is expressed on the same cell. Binding of such a bispecific mutein in a bicellular manner may result in mutual cross-linking of the CD95 apoptose receptor and the second cell surface receptor target antigen, which can effectively induce apoptosis of the cells (cf. Jung, G. et al. (2001) Cancer Res. 61, 1846-1848). However such a bispecific mutein may also have only binding affinity for one given target. Such a mutein may be useful as a molecular storage for drugs that are to be slowly released into the blood stream.
The term "mutagenesis" as used herein means that the experimental conditions are chosen such that the amino acid naturally occurring at a given sequence position of the lipocalin used can be substituted by at least one amino acid that is not present at this specific position in the respective natural polypeptide sequence. The term "mutagenesis" also includes the (additional) modification of the length of sequence segments by deletion or insertion of one or more amino acids. Thus, it is within the scope of the invention that, for example, one amino acid at a chosen sequence position is replaced by a stretch of three random mutations, leading to an insertion of two amino acid residues compared to the length of (the respective segment) of the wild-type protein. The term "random mutagenesis" refers to that no predetermined single amino acid (mutation) is present at a certain sequence position but that at least two amino acids can be incorporated into a selected sequence position during mutagenesis with a certain probability.
Such experimental conditions can, for example, be achieved by incorporating codons with a degenerate base composition into a nucleotide acid encoding the respective lipocalin employed. For example, use of the codon NNK or NNS (wherein N = adenine, guanine or cytosine or thymine; K = guanine or thymine; S = adenine or cytosine) allows incorporation of all 20 amino acids plus the amber stop codon during mutagenesis, whereas the codon VVS limits the number of possibly incorporated amino acids to 12, since it excludes the amino acids Cys, He, Leu, Met, Phe, Tip, Tyr, Val from being incorporated into the selected position of the polypeptide sequence; use of the codon NMS (wherein M = adenine or cytosine), for example, restricts the number of possible amino acids to 11 at a selected sequence position since it excludes the amino acids Arg,
Cys, Gly, He, Leu, Met, Phe, Trp, Val from being incorporated at a selected sequence position. In this respect it is noted that codons for other amino acids (than the regular 20 naturally occurring amino acids) such as selenocystein or pyrrolysine can also be incorporated into a nucleic acid of a mutein. It is also possible to use as described by Wang, L., et al., Science, 292:498-500, 2001 or Wang, L., Schultz, P.G., Chem. Comm., 1:1-11, 2002 "artificial" codons such as UAG which are usually recognized as stop codons in order to insert other unusual amino acids, for example O- methyl-L-tyrosine or p-aminophenylalanine.
The term "tear lipocalin" as used herein is not limited to the human tear lipocalin (SWISS-PROT Data Bank Accession Number M90424) but is meant to include all polypeptides having the structurally conversed lipocalin fold and a sequence homology with respect to the amino acid sequence of the human tear lipocalin of at least 60% . The term lipocalin fold is used in its regular meaning as used, e.g., in Flower, D.R. (1996), supra, to describe the typical three-dimensional lipocalin structure with a conformationally conserved β-barrel as a central motif made of a cylindrically closed β-sheet of eight antiparallel strands, wherein the open end of the barrel the β- strands are connected by four loops in a pairwise manner so that the binding pocket is formed (see also Fig. 4).
The definition of the peptide loops as used in the present invention is also in accordance with the regular meaning of the term lipocalin fold and is as follows and also illustrated in Fig. 4: The peptide loop (segment) AB connects the β-strands A and B of the cylindrically closed β-sheet, the peptide loop CD connects the β-strands C and D, the peptide loop EF connects the β-strands E and F, the peptide loop GH connects the β-strands G and H, the peptide loop BC connects the β- strands B and C, the loop DE connects the β-strands D and E, and the loop FG connects the β- strands F and G. As can be seen from Fig. 4 the loops AB, CD, EF and GH form the known binding site of the lipocalins (which was therefore called the open end), whereas, as found in the present invention, the loops BC, DE and FG can be used together with the N-terminal peptide stretch to form a second binding site which is located at the closed end of the β-barrel.
In accordance with the above, the term "tear lipocalin" includes structural homologues, already identified or yet to be isolated, from other species which have an amino acid sequence homology of more than about 60 %. The term "homology" as used herein in its usual meaning and includes identical amino acids as well as amino acids which are regarded to be conservative substitutions (for example, exchange of a glutamate residue by a aspartate residue) at equivalent positions in the linear amino acid sequence of two proteins that are compared with each other.
The percentage of homology is determined herein using the program BLASTP, version blastp 2.2.5 (November 16, 2002; cf. Altschul, S. F. et al. (1997) Nucleic Acids Res. 25, 3389-3402). The percentage of homology is based on the alignment of the entire polypeptide sequences (cutoff value set to 10" ) including the propeptide sequences, using the human tear lipocalin as reference in a pairwise comparison. It is calculated as the percentage of numbers of "positives" (homologues amino acids) indicated as result in the BLASTP program output divided by the total number of amino acids selected by the program for the alignment. It is noted in this connection that this total number of selected amino acids can differ from the length of the tear lipocalin (176 amino acids including the propeptide) as it is seen in the following. Examples of homologues proteins are Von Ebners gland protein 1 of Rattus norvegicus (VEGP protein; SWISS-PROT Data Bank Accession Numbers P20289) with a sequence homology of ca. 70 % (125 positives/178 positions including the propeptide; when the 18 residues long propeptides containing 13 "positives" are not taken into account: 112 positives/160, resulting also in an homology of ca.70 %), Von Ebners gland protein 2 of Rattus norvegicus (VEG protein 2; SWISS-PROT Data Bank Accession Numbers P41244) with a sequence homology of ca. 71 % (127 positives/178 including the propeptide; when the 18 residues long propeptides are not taken into account: 114 positives/160, the homology is determined to be also ca. 71 %), Von Ebners gland protein 2 of Sus scrofra (pig) (LCNl; SWISS-PROT Data Bank Accession Numbers P53715) with a sequence homology of about 74 % (131 positives/ 176 positions including the propeptide; when the 18 residues long propeptides containing 16 "positives" are not taken into account: 115 positives/158, resulting in an homology of ca. 73,0 %), or the Major allergen Can f 1 precursor of dog (ALL 1, SWISS-PROT Data Bank Accession Numbers 018873) with a sequence homology of ca.70.0 %, (122 positives/174 positions, or 110 positives/156 = ca. 70 % homology, when the propeptides with 12 positives are excluded) as determined with the program BLASTP as explained above. Such a structural homologue of the tear lipocalin can be derived from any species, i.e. from prokaryotic as well as from eukaryotic organisms. In case of eukaryotic organisms, the structural homologue can be derived from invertebrates as well as vertebrates such as mammals (e.g., human, monkey, dog, rat or mouse) or birds or reptiles.
In case a protein other than tear lipocalin is used in the present invention, the definition of the mutated sequence positions given for tear lipocalin can be assigned to the other lipocalin with the help of published sequence alignments or alignments methods which are available to the skilled artisan. A sequence alignment can, for example, be carried out as explained in WO 99/16873 (cf. Fig. 3 therein), using an published alignment such as the one in Fig. 1 of Redl, B. (2000) Biochim. Biophys. Acta, 1482, 241-248. If the three-dimensional structure of the lipocalins are available structural superpositions can also be used for the determination of those sequence positions that are to be subjected to mutagenesis in the present invention. Other methods of structural analysis such as multidimensional nuclear magnetic resonance spectroscopy can also be employed for this purpose.
The homologue of tear lipocalin can also be a mutein protein of tear lipocalin itself, in which amino acid substitutions are introduced at positions other than the positions selected in the present invention. For example, such a mutein can be a protein in which positions at the solvent exposed surface of the β-barrel are mutated compared to the wild type sequence of the tear lipocalin in order to increase the solubility or the stability of the protein.
In general, the term "tear lipocalin" includes all proteins that have a sequence homology of more than 60%, 70% 80%, 85%, 90% or 95% in relation to the human tear lipocalin (SWISS-PROT Data Bank Accession Number M90424).
In one preferred embodiment of the invention the mutein as disclosed herein is derived from human tear lipocalin. In other preferred embodiments the mutein is derived from the VEGP protein, VEG protein 2, LCN 1 or ALL 1 protein.
If the binding site at the closed end of the β-barrel is used, the mutein according to the invention typically comprises mutations at any two or more of the sequence positions in the peptide segments corresponding to the sequence positions 7-14, 41-49, 69-77, and 87-98 of the linear polypeptide sequence of human tear lipocalin. The positions 7-14 are part of the N-terminal peptide stretch, the positions 41-49 are comprised in the BC loop, the positions 60-77 are comprised in the DE loop and the positions 87-98 are comprised in the FG loop.
In mores specific embodiments of those muteins the mutations are introduced at those sequence positions which correspond to the positions 8, 9, 10, 11, 12, 13, 43, 45, 47, 70, 72, 74, 75, 90, 92, 94, and 97 of human tear lipocalin. Usually, such a mutein comprises mutations at 5-10 or 12-16 or all 17 of the sequence positions.
In case the binding site at the open end of the β-barrel is subjected to mutagenesis a lipocalin mutein according to the invention comprises mutations at any two or more of the sequence positions in the peptide segments corresponding to the sequence positions 24-36, 53-66, 79-84, and 103-110 of the linear polypeptide sequence of human tear lipocalin. The positions 24-36 are comprised in the AB loop, the positions 53-66 are comprised in the CD loop, the positions 69-77 are comprised in the EF loop and the positions 103-110 are comprised in the GH loop.
Accordingly, a mutein having two binding sites comprises mutations at any two or more of the sequence positions in the peptide segments corresponding to the sequence positions 7-14, 41-49, 69-77, and 87-97 of the linear polypeptide sequence of human tear lipocalin and additional mutations at any two or more of the sequence positions in the peptide segments corresponding to the sequence positions 24-36, 53-66, 79-84, and 103-110 of the linear polypeptide sequence of human tear lipocalin.
In this respect it is noted that the number of the segments (loops) defined above which are used for mutagenesis can vary (the N-terminal peptide stretch is included in the meaning of the term segment or loop). It is not necessary to mutate all four of these segments altogether of each of two binding sites, for example in a concerted mutagenesis. But it is also possible to introduce mutations only in one, two or three segments of each binding site in order to generate a mutein having detectable affinity to a given target. Therefore, it is possible to subject, for example, only two or three segments at the closed end of the β-barrel to mutagenesis if a binding molecule with only one engineered binding site is wanted. If this molecule is then wanted to have binding affinity towards a second target, sequence positions in any of the four loops of the second binding site can then be mutated.
The lipocalin muteins of the invention may comprise the wild-type (natural) amino acid sequence outside the mutated segments. On the other hand, the lipocalin muteins disclosed herein may also contain amino acid mutations outside the sequence positions subjected to mutagenesis as long as those mutations do not interfere with the binding activity and the folding of the mutein. Such mutations can be accomplished very easily on DNA level using established standard methods (Sambrook, J. et al. (1989) Molecular Cloning: A Laboratory Manual, 2nd Ed., Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY). Possible alterations of the amino acid sequence are insertions or deletions as well as amino acid substitutions. Such substitutions may be conservative, i.e. an amino acid residue is replaced with a chemically similar amino acid residue. Examples of conservative substitutions are the replacements among the members of the following groups: 1) alanine, serine, and threonine; 2) aspartic acid and glutamic acid; 3) asparagine and glutamine; 4) arginine and lysine; 5) isoleucine, leucine, methionine, and valine; and 6) phenylalanine, tyrosine, and tryptophan. One the other hand, it is also possible to introduce non-conservative alterations in the amino acid sequence.
Such modifications of the amino acid sequence include directed mutagenesis of single amino acid positions in order to simplify sub-cloning of the mutated lipocalin gene or its parts by incorporating cleavage sites for certain restriction enzymes. Furthermore, mutations can be introduced in order to improve certain characteristics of the mutein. The lipocalin muteins of the invention are able to bind the desired target with detectable affinity, i.e. with an affinity constant of preferably at least 105 M"1. Lower affinities are generally no longer measurable with common methods such as ELIS A and therefore of secondary importance. Especially preferred are lipocalin muteins, which bind the desired target with an affinity of at least 106 M_1, corresponding to a dissociation constant of the complex of 1 μM. The binding affinity of a mutein to the desired target can be measured by a multitude of methods such as fluorescence titration, competition ELIS A or surface plasmon resonance.
It is clear to the skilled person that complex formation is dependent on many factors such as concentration of the binding partners, the presence of competitors, ionic strength of the buffer system etc. Selection and enrichment is generally performed under conditions allowing the isolation of lipocalin muteins having an affinity constant of at least 105 M"1 to the target. However, the washing and elution steps can be carried out under varying stringency. A selection with respect to the kinetic characteristics is possible as well. For example, the selection can be performed under conditions, which favor complex formation of the target with muteins that show a slow dissociation from the target, or in other words a low k0f rate.
A tear lipocalin mutein of the invention may be used for complex formation with a given target. The target (ligand) may be any chemical compound in free or conjugated form which exhibits features of an immunological hapten, a hormone such as steroid hormones or any biopolymer or fragment thereof, for example, a protein or protein domain, a peptide, an oligodeoxynucleotide, a nucleic acid, an oligo- or polysaccharide or conjugates thereof. In a preferred embodiment of the invention the target is a protein. The protein can be any globular soluble protein or a receptor protein, for example, a trans-membrane protein involved in cell signaling, a component of the immune systems such as an MHC molecule or cell surface receptor that is indicative of a specific disease. The mutein may also be able to bind only fragments of a protein. For example, a mutein can bind to a domain of a cell surface receptor, when it is part of the receptor anchored in the cell membrane as well as to the same domain in solution, if this domain can be produced as a soluble protein as well. However the invention is by no means limited to muteins that only bind such macromolecular targets. But it is also possible to obtain muteins of tear lipocalin by means of mutagenesis which show specific binding affinity to ligands of low(er) molecular weight such as biotin, fluorescein or digoxigenin.
For some applications, it is useful to employ the muteins of the invention in a labeled form. Accordingly, the invention is also directed to lipocalin muteins which are conjugated to a label selected from the group consisting of enzyme labels, radioactive labels, colored labels, fluorescent labels, chromogenic labels, luminescent labels, haptens, digoxigenin, biotin, metal complexes, metals, and colloidal gold. The mutein may also be conjugated to an organic molecule. The term "organic molecule" as used herein preferably denotes an organic molecule comprising at least two carbon atoms, but preferably not more than seven rotatable carbon bonds, having a molecular weight in the range between 100 and 2000 Dalton, preferably 1000 Dalton, and optionally including one or two metal atoms.
In general, it is possible to label the lipocalin mutein with any appropriate chemical substance or enzyme, which directly or indirectly generates a detectable compound or signal in a chemical, physical or enzymatic reaction. An example for a physical reaction is the emission of fluorescence upon irradiation or the emission of X-rays when using a radioactive label. Alkaline phosphatase, horseradish peroxidase or β-galactosidase are examples of enzyme labels which catalyze the formation of chromogenic reaction products. In general, all labels commonly used for antibodies (except those exclusively used with the sugar moiety in the Fc part of immunoglobulins) can also be used for conjugation to the muteins of the present invention. Such conjugates can be produced by methods well known in the art.
For several applications of the muteins disclosed herein it may be advantageous to use them in the form of fusion proteins . In preferred embodiments the inventive lipocalin mutein is fused at its N- terminus or its C-terminus to a protein, a protein domain or a peptide such as a signal sequence and/or an affinity tag.
The fusion partner may confer new characteristics to the inventive hpocalin mutein such as enzymatic activity or binding affinity for other molecules. Examples of suitable fusion proteins are alkaline phosphatase, horseradish peroxidase, gluthation-S-transferase, the albumin-binding domain of protein G, protein A, antibody fragments, oligomerization domains, lipocalin muteins of same or different binding specificity (which results in the formation of "duocahns", cf. Schlehuber, S. & Skerra, A. (2001), Biol. Chem. 382, 1335-1342), or toxins. If two bispecific lipocalin mutein of the inventions (i.e. each of them has two binding sites) are combined into a
"duocalin", a tetravalent molecule is formed. If for example a duocalin is generated from only one mutein having two binding sites that specifically bind biotin, a tetravalent molecule (homodimer) comparable to streptavidin (which is a homotetramer, in which each monomer binds one biotin molecule) can be obtained. Due to expected avidity effects such a mutein might be a useful analytical tools in methods that make use of the detection of biotin groups. Affinity tags such as the Strep-tag® or Strep-tag® II (Schmidt, T.G.M. et al. (1996) J. Mol. Biol. 255, 753-766), the myc-tag, the FLAG-tag, the His6-tag or the HA-tag or proteins such as glutathione-S-transf erase also allow easy detection and/or purification of recombinant proteins are further examples of preferred fusion partners. Finally, proteins with chromogenic or fluorescent properties such as the green fluorescent protein (GFP) or the yellow fluorescent protein (YFP) are suitable fusion partners for a lipocalin mutein of the invention as well.
The term "fusion protein" as used herein also comprises lipocalin muteins according to the invention containing a signal sequence. Signal sequences at the N-terminus of a polypeptide direct this polypeptide to a specific cellular compartment, for example the periplasm of E. coli or the endoplasmatic reticulum of eukaryotic cells. A large number of signal sequences is known in the art. A preferred signal sequence for secretion a polypeptide into the periplasm of E. coli is the OmpA-signal sequence.
The present invention also relates to nucleic acid molecules (DNA and RNA) comprising nucleotide sequences coding for muteins as described herein. Since the degeneracy of the genetic code permits substitutions of certain codons by other codons specifying the same amino acid, the invention is not limited to a specific nucleic acid molecule encoding a fusion protein of the invention but includes all nucleic acid molecules comprising nucleotide sequences encoding a functional fusion protein.
In one preferred embodiment of the nucleic acid molecule of invention its sequence is derived from the coding sequence of human tear lipocalin. In other preferred embodiments the nucleic acid is derived from the VEGP protein, VEG protein 2, LCN 1 or ALL 1 protein
In another preferred embodiment the nucleic acid sequence encoding a mutein according to the invention comprises mutations at any two or more of the sequence positions in the peptide segments corresponding to the sequence positions 7-14, 43-49, 70-77, and 87-97 of the linear polypeptide sequence of human tear lipocalin, with the sequence positions corresponding to the positions 8, 9, 10, 11, 12, 13, 43, 45, 47, 70, 72, 74, 75, 90, 92, 94, and 97 ofhuman tear lipocalin being particularly preferred.
In a further preferred embodiment the nucleic acid sequence encoding a mutein according to the invention comprises mutations at any two or more of the sequence positions in the peptide segments corresponding to the sequence positions 24-36, 53-66, 79-84, and 103-110 of the linear polypeptide sequence of human tear lipocalin.
Also preferred are nucleic acid molecules encoding a mutein of the invention comprising mutations at any two or more of the sequence positions in the peptide segments corresponding to the sequence positions 7-14, 43-49, 70-77, and 87-97 of the linear polypeptide sequence of human tear lipocalin mutations and additional mutations at any two or more of the sequence positions in the peptide segments corresponding to the sequence positions 24-36, 53-66, 79-84, and 103-110 of the linear polypeptide sequence of human tear lipocalin.
The invention as disclosed herein also includes nucleic acid molecules encoding TLPC muteins, which comprise additional mutations outside the segments of experimental mutagenesis. Such mutations are often tolerated or can even prove to be advantageous, for example if they contribute to an improved folding efficiency, protein stability or ligand binding affinity of the mutein.
A nucleic acid molecule disclosed in this application may be "operably linked" to a regulatory sequence (or regulatory sequences) to allow expression of this nucleic acid molecule.
A nucleic acid molecule, such as DNA, is referred to as "capable of expressing a nucleic acid molecule" or capable "to allow expression of a nucleotide sequence" if it comprises sequence elements which contain information regarding to transcriptional and/or translational regulation, and such sequences are "operably linked" to the nucleotide sequence encoding the polypeptide. An operable linkage is a linkage in which the regulatory sequence elements and the sequence to be expressed are connected in a way that enables gene expression. The precise nature of the regulatory regions necessary for gene expression may vary among species, but in general these regions comprise a promoter which, in prokaryotes, contains both the promoter per se, i.e. DNA elements directing the initiation of transcription, as well as DNA elements which, when transcribed into RNA, will signal the initiation of translation. Such promoter regions normally include 5 ' non-coding sequences involved in initiation of transcription and translation, such as the -35/-10 boxes and the Shine-Dalgarno element in prokaryotes or the TATA box, CAAT sequences, and 5 -capping elements in eukaryotes. These regions can also include enhancer or repressor elements as well as translated signal and leader sequences for targeting the native polypeptide to a specific compartment of a host cell. In addition, the 3' non-coding sequences may contain regulatory elements involved in transcriptional termination, polyadenylation or the like. If, however, these termination sequences are not satisfactory functional in a particular host cell, then they may be substituted with signals functional in that cell.
Therefore, a nucleic acid molecule of the invention can include a regulatory sequence, preferably a promoter sequence. In another preferred embodiment, a nucleic acid molecule of the invention comprises a promoter sequence and a transcriptional termination sequence. Suitable prokaryotic promoters are, for example, the tet promoter, the ZαcUV5 promoter or the T7 promoter. Examples of promoters useful for expression in eukaryotic cells are the SV40 promoter or the CMV promoter.
The nucleic acid molecules of the invention can also be comprised in a vector or any other cloning vehicles, such as plasmids, phagemids, phage, baculovirus, cosmids or artificial chromosomes. In a preferred embodiment, the nucleic acid molecule is comprised in a phasmid. A phasmid vector denotes a vector encoding the intergenic region of a temperent phage, such as M13 or f 1, or a functional part thereof fused to the cDNA of interest. After superinfection of the bacterial host cells with such an phagemid vector and an appropriate helper phage (e.g. M13K07, VCS-M13 or R408) intact phage particles are produced, thereby enabling physical coupling of the encoded heterologous cDNA to its corresponding polypeptide displayed on the phage surface
(reviewed, e.g., in Kay, B.K. et al. (1996) Phage Display ofPeptides and Proteins - A Laboratory Manual, 1st Ed., Academic Press, New York NY; Lowman, H. B. (1997) Annu. Rev. Biophys. Biomol. Struct. 26, 401-424 or Rodi, D. J. & Makowski, L. (1999) Curr. Opin. Biotechnol. 10, 87-93).
Such cloning vehicles can include, aside from the regulatory sequences described above and a nucleic acid sequence encoding a lipocalin mutein of the invention, replication and control sequences derived from a species compatible with the host cell that is used for expression as well as selection markers conferring a selectable phenotype on transformed or transfected cells. Large numbers of suitable cloning vectors are known in the art, and are commercially available.
The DNA molecule encoding lipocalin muteins of the invention, and in particular a cloning vector containing the coding sequence of such a lipocalin mutein can be transformed into a host cell capable of expressing the gene. Transformation can be performed using standard techniques (Sambrook, J. et al. (1989), supra). Thus, the invention is also directed to a host cell containing a nucleic acid molecule as disclosed herein.
The transformed host cells are cultured under conditions suitable for expression of the nucleotide sequence encoding a fusion protein of the invention. Suitable host cells can be prokaryotic, such as Escherichia coli (E. coli) or Bacillus subtilis, or eukaryotic, such as Saccharomyces cerevisiae, Pichiapastoris, SF9 or High5 insect cells, immortalized mammalian cell lines (e.g. HeLa cells or CHO cells) or primary mammalian cells.
The invention also relates to a method for the generation of a mutein according to the invention or a fusion protein thereof, comprising: (a) subjecting a nucleic acid molecule encoding a tear lipocalin of a functional homologue thereof, wherein said tear lipocalin or functional homologue thereof has at least 60% sequence homology with human tear lipocalin, to mutagenesis at two or more different nucleotide triplets, resulting in one or more mutein nucleic acid molecules(s); (b) expressing the one or more mutein nucleic acid molecule(s) obtained in (a) in a suitable expression system, and (c) enriching at least one mutein having a detectable binding affinity for a given target by means of selection and/or isolation.
In further embodiments of this method, the nucleic acid molecule can be individually subjected to mutagenesis at two or more different nucleotide triplets in any one, two, three or all four above-mentioned peptide segments arranged at either end of the β-barrel structure.
In the method of generation a mutein or a fusion protein thereof is obtained starting from the nucleic acid encoding tear lipocalin or a functional homologue thereof, which is subjected to mutagenesis and introduced into a suitable bacterial or eukaryotic host organism by means of recombinant DNA technology (as already outlined above).
The coding sequence of, for example, human tear lipocalin (Redl, B. et al. (1992) /. Biol. Chem. 267, 20282-20287) can serve as a starting point for mutagenesis of the peptide segments selected in the present invention. For the mutagenesis of the amino acids in the N-terminal peptide stretch and the three peptide loops BC, DE, and FG at the end of the β-barrel structure that is located opposite of the natural lipocalin binding pocket as well as the four peptide loops AB, CD, EF, and GH encompassing said binding pocket, the person skilled in the art has at his disposal the various established standard methods for site-directed mutagenesis (Sambrook, J. et al. (1989), supra). A commonly used technique is the introduction of mutations by means of PCR (polymerase chain reaction) using mixtures of synthetic oligonucleotides, which bear a degenerate base composition at the desired sequence positions. The use of nucleotide building blocks with reduced base pair specificity, as for example inosine, is another option for the introduction of mutations into a chosen sequence segment. A further possibility is the so-called triplet-mutagenesis. This method uses mixtures of different nucleotide triplets each of which codes for one amino acid for the incorporation into the coding sequence.
One possible strategy for introducing mutations in the selected regions of the respective polypeptides is based on the use of four oligonucleotides, each of which is partially derived from one of the corresponding sequence segments to be mutated (cf. Fig.3). When synthesizing these oligonucleotides, a person skilled in the art can employ mixtures of nucleic acid building blocks for the synthesis of those nucleotide triplets which correspond to the amino acid positions to be mutated so that codons encoding all natural amino acids randomly arise, which at last results in the generation of a lipocalin peptide library. For example, the first oligonucleotide corresponds in its sequence - apart from the mutated positions - to the coding strand for the peptide segment to be mutated at the most N-terminal position of the lipocalin polypeptide. Accordingly, the second oligonucleotide corresponds to the non-coding strand for the second sequence segment following in the polypeptide sequence. The third oligonucleotide corresponds in turn to the coding strand for the corresponding third sequence segment. Finally, the fourth oligonucleotide corresponds to the non-coding strand for the fourth sequence segment. A polymerase chain reaction can be performed with the respective first and second oligonucleotide and separately, if necessary, with the respective third and fourth oligonucleotide.
The amplification products of both of these reactions can be combined by various known methods into a single nucleic acid comprising the sequence from the first to the fourth sequence segments, in which mutations have been introduced at the selected positions. To this end, both of the products can for example be subjected to a new polymerase chain reaction using flanking oligonucleotides as well as one or more mediator nucleic acid molecules, which contribute the sequence between the second and the third sequence segment. This procedure is schematically reproduced in Fig. 3. In the choice of the number and arrangement within the sequence of the oligonucleotides used for the mutagenesis, the person skilled in the art has numerous alternatives at his disposal. The nucleic acid molecules defined above can be connected by ligation with the missing 5 - and 3 -sequences of a nucleic acid encoding a lipocalin polypeptide and/or the vector, and can be cloned in a known host organism. A multitude of established procedures are available for ligation and cloning (Sambrook, J. et al. (1989), supra). For example, recognition sequences for restriction endonucleases also present in the sequence of the cloning vector can be engineered into the sequence of the synthetic oligonucleotides. Thus, after amplification of the respective PCR product and enzymatic cleavage the resulting fragment can be easily cloned using the corresponding recognition sequences.
Longer sequence segments within the gene coding for the protein selected for mutagenesis can also be subjected to random mutagenesis via known methods, for example by use of the polymerase chain reaction under conditions of increased error rate, by chemical mutagenesis or by using bacterial mutator strains. Such methods can also be used for further optimization of the target affinity or specificity of a lipocalin mutein. Mutations possibly occurring outside the segments of experimental mutagenesis are often tolerated or can even prove to be advantageous, for example if they contribute to an improved folding efficiency or folding stability of the lipocalin mutein.
After expression of the nucleic acid sequences that were subjected to mutagenesis in an appropriate host, the clones carrying the genetic information for the plurality of respective lipocalin muteins, which bind a given target can be selected from the library obtained. Well known techniques can be employed for the selection of these clones, such as phage display (reviewed in Kay, B.K. et al. (1996) supra; Lowman, H. B. (1997) supra or Rodi, D. J. & Makowski, L. (1999) supra), colony screening (reviewed in Pini, A. et al. (2002) Comb. Chem.
High Throughput Screen. 5, 503-510), ribosome display (reviewed in Amstutz, P. et al. (2001) Curr. Opin. Biotechnol 12, 400-405) or mRNA display as reviewed in Wilson, D.S. et al. (2001) Proc. Natl. Acad. Sci. USA 98, 3750-3755.
An embodiment of the phage display technique (reviewed in Kay, B.K. et al. (1996) supra; Lowman, H. B. (1997) supra or Rodi, D. J. & Makowski, L. (1999) supra) using temperent M13 phage is given as an example of a selection method according to the invention. However, it is noted that other temperent phage such as f 1 or lytic phage such as T7 may be employed as well. For the exemplary selection method, M13 phagemids (cf . also above) are produced which allow the expression of the mutated lipocalin nucleic acid sequence as a fusion protein with a signal sequence at the N-terminus, preferably the OmpA-signal sequence, and with the capsid protein pHI of the phage M13 or fragments thereof capable of being incorporated into the phage capsid at the C-terminus. The C-terminal fragment ΔpIII of the phage capsid protein comprising amino acids 217 to 406 of the wild-type sequence is preferably used to produce the fusion proteins. Especially preferred is a C-terminal fragment of pIJJ, in which the cysteine residue at position 201 is missing or is replaced by another amino acid.
The fusion protein may comprise additional components such as an affinity tag, which allows the immobilization and/or purification of the fusion protein or its parts. Furthermore, a stop codon can be located between the sequence regions encoding the lipocalin or its muteins and the phage capsid gene or fragments thereof, wherein the stop codon, preferably an amber stop codon, is at least partially translated into an amino acid during translation in a suitable suppressor strain.
For example, the phagemid vector pTLPC7 (Fig. 1) can be used for the construction of a phage library encoding human tear lipocalin muteins. The inventive nucleic acid molecules coding for the mutated peptide segments are inserted into the vector using the BstXI restriction sites. Recombinant vectors are then transformed into a suitable host strain such as E. coli XLl-Blue. The resulting library is subsequently superinfected in liquid culture with an appropriate M13- helper phage in order to produce functional phage. The recombinant phagemid displays the lipocalin mutein on its surface as a fusion with the coat protein pIJJ or a fragment thereof, while the N-terminal signal sequence of the fusion protein is normally cleaved off. On the other hand, it also bears one or more copies of the native capsid protein pIJJ supplied by the helper phage and is thus capable of infecting a recipient, in general a bacterial strain carrying a F- or F'-plasmid. During or after infection gene expression of the fusion protein comprised of the lipocalin mutein and the capsid protein pill can be induced, for example by addition of anhydrotetracycline. The induction conditions are chosen such that a substantial fraction of the phage obtained displays at least one lipocalin mutein on their surface. Various methods are known for isolating the phage, such as precipitation with polyethylene glycol. Isolation typically occurs after an incubation period of 6-8 hours.
The isolated phage are then subjected to a selection process by incubating them with a given target, wherein the target is present in a form allowing at least a temporary immobilization of those phage displaying muteins with the desired binding activity. Several immobilization methods are known in the art. For example, the target can be conjugated with a carrier protein such as serum albumin and be bound via this carrier to a protein-binding surface such as polystyrene. Microtiter plates suitable for ELISA techniques or so-called "immunosticks" are preferred. Alternatively, conjugates of the target can also be implemented with other binding groups such as biotin. The target can then be immobilized on surfaces, which will selectively bind this group, such as microtiter plates or paramagnetic particles coated with avidin or streptavidin.
For example, the phage particles are captured by binding to the respective target immobilized on the surface. Unbound phage particles are subsequently removed by iterative washing. For the elution of bound phage, free target (ligand) molecules can be added to the samples as a competitor. Alternatively, elution can also be achieved by adding proteases or under moderately denaturing conditions, e.g. in the presence of acids, bases, detergents or chaotropic salts. A preferred method is the elution using buffers having pH 2.2, followed by neutralization of the solution. The eluted phage may then be subjected to another selection cycle. Preferably, selection is continued until at least 0.1 % of the clones comprise lipocalin muteins with detectable affinity for the respective target. Depending on the complexity of the library employed 2-8 cycles are required to this end.
For the functional analysis of the selected lipocalin muteins, an E. coli host strain is infected with the phagemids obtained and phagemid DNA is isolated using standard techniques (Sambrook, J. et al. (1989), supra). The mutated sequence fragment or the entire lipocalin mutein nucleic acid sequence can be sub-cloned in any suitable expression vector. The recombinant lipocalin muteins obtained can be purified from their host organism or from a cell lysate by various methods known in the art such as gel filtration or affinity chromatography.
However, the selection of lipocalin muteins can also be performed using other methods well known in the art. Furthermore, it is possible to combine different procedures. For example, clones selected or at least enriched by phage display can subsequently be subjected to a colony-screening assay in order to directly isolate a particular lipocalin mutein with detectable binding affinity for a given target. Additionally, instead of generating a single phage library comparable methods can be applied in order to optimize a mutein with respect to its affinity or specificity for the desired target by repeated, optionally limited mutagenesis of its coding nucleic acid sequence.
The invention also relates to a method for the production of a mutein of the invention, wherein the mutein, a fragment of the mutein or a fusion protein of the mutein and another polypeptide is produced starting from the nucleic acid coding for the mutein by means of genetic engineering methods. The method can be carried out in vivo, the mutein can for example be produced in a bacterial or eucaryotic host organism and then isolated from this host organism or its culture. It is also possible to produce a protein in vitro, for example by use of an in vitro translation system.
When producing the mutein in vivo a nucleic acid encoding a mutein of the invention is introduced into a suitable bacterial or eukaryotic host organism by means of recombinant DNA technology (as already outlined above). For this purpose, the host cell is first transformed with a cloning vector comprising a nucleic acid molecule encoding a mutein of the invention using established standard methods (Sambrook, J. et al. (1989), supra). The host cell is then cultured under conditions, which allow expression of the heterologous DNA and thus the synthesis of the corresponding polypeptide. Subsequently, the polypeptide is recovered either from the cell or from the cultivation medium. Since many lipocalins comprise intramolecular disulfide bonds, it can be preferred to direct the polypeptide to a cell compartment having an oxidizing redox-milieu using an appropriate signal sequence. Such an oxidizing environment is provided in the periplasm of Gram-negative bacteria such as E. coli or in the lumen of the endoplasmatic reticulum of eukaryotic cells and usually favors the correct formation of the disulfide bonds. It is, however, also possible to generate a mutein of the invention in the cytosol of a host cell, preferably E. coli. In this case, the polypeptide can, for instance, be produced in form of inclusion bodies, followed by renaturation in vitro. A further option is the use of specific host strains having an oxidizing intracellular milieu, which thus allow the production of the native protein in the cytosol.
However, a mutein of the invention may not necessarily be generated or produced only by use of genetic engineering. Rather, a lipocalin mutein can also be obtained by chemical synthesis such as Merrifield solid phase polypeptide synthesis. It is for example possible that promising mutations are identified using molecular modeling and then to synthesize the wanted (designed) polypeptide in vitro and investigate the binding activity for a given target.
The invention also relates to a pharmaceutical composition comprising at least one inventive mutein or a fusion protein thereof and a pharmaceutically acceptable excipient.
The lipocalin muteins according to the invention can be administered via any parenteral or non- parenteral (enteral) route that is therapeutically effective for proteinaceous drugs. Parenteral application methods comprise, for example, intracutaneous, subcutaneous, intramuscular or intravenous injection and infusion techniques, e.g. in the form of injection solutions, infusion solutions or tinctures, as well as aerosol installation and inhalation, e.g. in the form of aerosol mixtures, sprays or powders. Non-parenteral delivery modes are, for instance, orally, e.g. in the form of pills, tablets, capsules, solutions or suspensions, or rectally, e.g. in the form of suppositories. The muteins of the invention can be administered systemically or topically in formulations containing conventional non-toxic pharmaceutically acceptable excipients or carriers, additives and vehicles as desired.
In a preferred embodiment of the present invention the pharmaceutical is administered parenterally to a mammal, and in particular to humans, with aerosol installation being the most preferable application method due to the low molecular weight of the muteins.
Accordingly, the muteins of the present invention can be formulated into compositions using pharmaceutically acceptable ingredients as well as established methods of preparation (Gennaro, A.L. and Gennaro, A.R. (2000) Remington: The Science and Practice of Pharmacy, 20th Ed., Lippincott Williams & Wilkins, Philadelphia, PA). To prepare the pharmaceutical compositions, pharmaceutically inert inorganic or organic excipients can be used. To prepare e.g. pills, powders, gelatin capsules or suppositories, for example, lactose, talc, stearic acid and its salts, fats, waxes, solid or liquid polyols, natural and hardened oils. Suitable excipients for the production of solutions, suspensions, emulsions, aerosol mixtures or powders for reconstitution into solutions or aerosol mixtures prior to use include water, alcohols, glycerol, polyols, and suitable mixtures thereof as well as vegetable oils.
The pharmaceutical composition may also contain additives, such as, for example, fillers, binders, wetting agents, glidants, stabilizers, preservatives, emulsifiers, and furthermore solvents or solubilizers or agents for achieving a depot effect. The latter is that fusion proteins may be incorporated into slow or sustained release or targeted delivery systems, such as liposomes and microcapsules.
The formulations can be sterilized by numerous means, including filtration through a bacteria- retaining filter, or by incorporating sterilizing agents in the form of sterile solid compositions which can be dissolved or dispersed in sterile water or other sterile medium just prior to use.
As is evident from the above disclosure, a mutein of the present invention or a fusion protein or a conjugate thereof can be employed in many applications. In general, such a mutein can be used in all applications antibodies are used, except those with specifically rely on the glycosylation of the Fc part. A mutein of the invention can also be used for the targeting of a compound to a preselected site. For such a purpose the mutein is contacted with the compound of interest in order to allow complex formation. Then the complex comprising the mutein and the compound of interest are delivered the preselected site. This use is in particular suitable, but not restricted to, for delivering a drug (selectively) to the site such an infected body part or organ which is supposed to be treated with the drug.
Another use of the inventive muteins is the binding/detection of a given target or target molecule, comprising contacting the mutein with a test sample supposed to contain said target, and detecting of the mutein/target complex by a suitable signal. A mutein can also be used for the separation of a given target, comprising contacting the mutein with a sample supposed to contain said target in order to allow complex formation, and separating the mutein/target complex from the sample. In such uses the complex comprising the mutein and the target may be immobilized on any suitable solid phase.
The detectable signal can be caused by a label, as explained above, or by a change of physical properties due to the binding, i.e. the complex formation, itself. One example is plasmon surface resonance, the value of which is changed during binding of binding partners from which one is immobilized on a surface such as a gold foil.
The muteins disclosed herein and its derivatives can thus be used in many fields similar to antibodies or fragments thereof. In addition to their use for binding to a support, allowing the target of a given mutein or a conjugate or a fusion protein of this target to be immobilized or separated, the muteins can be used for labeling with an enzyme, an antibody, a radioactive substance or any other group having biochemical activity or defined binding characteristics. By so doing, their respective targets or conjugates or fusion proteins thereof can be detected or brought in contact with them. For example, muteins of the invention can serve to detect chemical structures by means of established analytical methods (e.g. ELISA or Western Blot) or by microscopy or immunosensorics. Here, the detection signal can either be generated directly by use of a suitable mutein conj ugate or fusion protein or indirectly by immunochemical detection of the bound mutein via an antibody.
Numerous possible applications for the inventive muteins also exist in medicine. In addition to their use in diagnostics and drug delivery, a mutant polypeptide of the invention, which binds, for example, tissue- or tumor-specific cellular surface molecules can be generated. Such a mutein may, for example, be employed in conjugated form or as a fusion protein for "tumor imaging" or directly for cancer therapy.
Another related and preferred use of a mutein described herein is target validation, i.e. the analysis whether a polypeptide assumed to be involved in the development or progress of a disease or disorder is indeed somehow causative of that disease or disorder. This use for validating a protein as a pharmacological drug target takes advantage of the ability of a mutein of the present invention to specifically recognize a surface area of a protein in its native conformation, i.e. to bind to a native epitope. In this respect, it is to be noted that this ability has been reported only for a limited number of recombinant antibodies. However, the use of an inventive mutein for validation of a drug target is not limited to the detection of proteins as targets, but also includes the detection of protein domains, peptides, nucleic acid molecules, organic molecules or metal complexes.
The invention is further illustrated by the following Figures and Examples in which.
Figure 1 schematically depicts the phagemid vector pTLPC7;
Figure 2 schematically depicts the phasmid vector pTLPC6;
Figure 3 schematically illustrates the generation of the library of tear lipocalin muteins at the nucleic acid level;
Figure 4 schematically depicts the structure of the lipocalin fold;
Figure 5 schematically depicts the expression vector pASK75-strepU-CD47;
Figure 6 shows the polypeptide sequence of mature human tear lipocalin (SWISS-PROT Data Bank Accession Number M90424); and
Figure 7 shows a schematic drawing of the expression vector pBBP46.
Fig.1 shows a schematic drawing of the vector pTLPC7 encoding a fusion protein comprised of the OmpA signal sequence (OmpA), a modified TLPC with the amino acid substitutions Ala5Asp, SerόGly, Asp7Gly, CyslOlSer, and Glul04Gln (for the TLPC cDNA, see Redl et al., supra) and a truncated form of the M13 coat protein pHJ, comprising amino acids 217 to 406 (pin). Gene expression is under the control of the tetracycline promoter/operator (tetP o) system. Transcription is terminated at the lipoprotein transcription terminator (tιpp). The vector further comprises an origin of replication (ori), the intergenic region of the filamentous phage f 1 (fl-IG), the ampicillin resistance gene (bla) coding for β-lactamase and the tetracycline repressor gene (tetR). An amber stop codon, which is partially translated into Gin in SupE amber suppressor host strain, is located between the TLPC coding region and the coding region for the truncated phage coat protein pffl. Both the JSstXI-restriction sites used for the cloning of the mutated gene cassette and the restriction sites flanking the structural gene are labeled. The nucleic acid sequence of a Xbal- H dπi segment of pTLPC7 is shown together with the encoded amino acid sequence in the sequence listing as SEQ ID NO: 1. The vector sequence outside this region is identical with that of pASK75, the complete nucleotide sequence of which is given in the German patent publication DE 44 17 598 Al.
Fig. 2 shows a schematic drawing of the vector pTLPC6. pTLPC6 encodes a fusion protein comprised of the OmpA signal sequence, a modified TLPC according to Fig. 1, and the Strep- tag® n affinity tag. Otherwise, the vector is identical to pTLPC7. The nucleic acid sequence of a Xbal- H diπ segment of pTLPC6 is shown together with the encoded amino acid sequence in the sequence listing as SEQ ID NO: 2. The vector sequence outside this region is identical with that of pASK75, the complete nucleotide sequence of which is given in the German patent publication DE 44 17 598 Al.
Fig. 3 schematically shows a strategy for the concerted mutagenesis of 17 selected amino acid positions in the modified TLPC by repeated polymerase chain reaction (PCR). For the sequence near the N-terminus as well as for each of the three peptide loops BC, DE, and FG, respectively, in which the amino acids are to be mutated, an oligodeoxynucleotide was synthesized, (SEQ ID NO: 3, SEQ ID NO: 4, SEQ ID NO: 5, and SEQ ID NO: 6), bearing random nucleotides as indicated in the sequence listing. Due to the composition chosen, from the altogether three possible stop codons only the amber stop codon, TAG, was allowed at the mutated codons, which is translated as glutamine in the E. coli supΕ strains XLl-blue (Bullock et al. (1987)
BioTechniques 5, 376-378) or TGI (Sambrook et al., supra). For certain applications, for example gene expression in other bacterial strains or organisms, such a nonsense codon can be substituted by a glutamine-encoding codon, e.g., by site-directed mutagenesis. A nucleic acid fragment with 159 base pairs was amplified (PCR No. 1, A) with the respective primers SΕQ ID NO: 3 and SΕQ ID NO: 4 using the pTLPC6 plasmid-DNA (SΕQ ID NO: 2) as a template. In another PCR, a nucleic acid fragment with 123 base pairs was amplified (PCR no. 1 , B) with the primers SEQ ID NO: 5 and SEQ ID NO: 6, respectively, also using pTLPC6 as template. The mixture of both PCR products served as a template in another amplification (PCR No. 2) with the two 5 - biotinylated flanking PCR primers, namely SEQ ID NO: 7 and SEQ ID NO: 8, and a mediating primer SEQ ID NO: 9, resulting in the amplification of a DNA fragment of 341 base pairs. This fragment comprising a mixture of all 17 mutated codons was subsequently cloned into the vector pTPLC7 using the two BstXI restriction sites, the special arrangement of which led to two non- compatible overhanging DNA ends enabling a particularly efficient ligation. The ligation efficiency could be improved by purification of the digested PCR-fragment by paramagnetic streptavidin coated beads. The amino acid substitution Glul04Gln as well as the silent mutations in the codon for Ala-3 of the ompA signal sequence, in the codon for Ala21 and Hisl06 were previously accomplished during the construction of pTLPC6 in order to introduce both of the BstXI restriction sites into the TLPC coding sequence.
Fig. 4 schematically illustrates the characteristic features of the lipocalin fold (according to Flower, D.R. (1996), supra). The eight β-strands of the antiparallel β-sheet which form the β- barrel) are shown as arrows and labeled A to H (a ninth β-strand, designated I which is additionally present in some lipocalins, is also schematically shown). The hydrogen-bonded connection of two strands is indicated by a pair of dotted lines between them. The connecting loops are shown as solid curved lines. The two ends of the β-barrel are topologically distinct. One end has four β-hairpins (loops AB, CD, EF and GH), the opening of the known ligand binding site of the lipocalins is here and called the open end. The other end of the β-barrel has three loops (BC, DE and FG) which together with the N-terminal polypeptide region build the closed end and are used in the present invention to introduce an alternative binding site. The parts which form the three main structurally conserved regions (SCRs) of the fold, SCRl , SCR2 and SCR3, are marked as boxes.
Fig. 5 shows a schematic drawing of the expression vector pASK75-strepn-CD47. pASK75- strepπ~CD47 codes for a fusion protein made of the OmpA signal sequence, a modified extracellular domain of human CD47 comprising amino acids 1 to 119 of the mature protein with the amino acid substitution Cys 15 to Ala and the Strep-tag® π affinity tag. All further genetic elements are identical with the generic vector pASK75. A relevant segment from the nucleic acid sequence of pASK75-strepH-CD47 is reproduced together with the encoded amino acid sequence in the sequence listing as SEQ ID NO: 10. The segment begins with the Xbal restriction site and ends with the H dUI restriction site. The vector elements outside this region are identical with the vector pASK75, the complete nucleotide sequence of which is exhibited in the German patent publication DE 44 17 598 Al.
Fig. 6 shows the polypeptide sequence of mature human tear lipocalin (SWISS-PROT Data Bank Accession Number M90424, 158 amino acids, cf. also Redl B. (2000) Biochim. Biophys. Acta, supra). In this respect it is noted that a human protein that was modified as follows was used in the following examples for the generation of lipocalin muteins. First, the first four N-terminal amino acid residues of the deposited sequence of human tear lipocalin (HHLL) were deleted. Second, the last two C-terminal amino acid residues (SD) were also deleted. Third, the wild type sequence at sequence positions 5 to 7 (ASD) was changed to GGD. These changes are reflected in the attached sequence listings, in which the amino acids GGD are indicated as first three residues of the used tear lipocalin. The four segments (AB, CD, EF and GH) at the open end of the β-barrel in which amino acids are exchanged are marked below the sequence of TLPC by double underlining. The segments BC, DE and FG as well as the N-terminal peptide stretch in which mutations are introduced to create a binding site at the closed end of the β-barrel are marked in bold and single underlining. The sequences position of TLPC which are mutated in the examples are additionally labelled with asterisks.
Fig.7 shows a schematic drawing of the expression vector pBBP46. pBBP46 codes for a fusion protein of the OmpA signal sequence and the T7 detection tag (T7) with a modified bilin binding protein of Pieris brassicae (cf. SEQ ID NO: 11) including the C-terminal Strep-tag® π. This structural gene is followed by the dsbC structural gene (including its ribosomal binding site) from E. coli (Zapun et al., Biochemistry 34 (1995), 5075-5089) as a second cistron (dsbC). A relevant segment of the nucleic acid sequence of pBBP46 is reproduced together with the encoded amino acid sequence in the sequence listing as SEQ ID NO: 11. The segment begins with the Xbal restriction site and ends with the HzπdlTl restriction site. The vector elements outside this region are identical with the vector pASK75, the complete nucleotide sequence of which is exhibited in the German patent publication DE 44 17 598 Al.
Examples
Example 1: Generation of a library with about 10 billion independent TLPC muteins
Unless otherwise indicated, established recombinant genetic methods were used, for example as described in Sambrook et al. (supra). A random library of TLPC with high complexity was prepared by concerted mutagenesis of in total 17 selected amino acid positions near the N-terminus and in the peptide loops BC, DE as well as FG using PCR in multiple steps according to Fig.3. The PCR reactions were performed in a volume of 100 μl in both of the first amplification steps (PCR No. 1 , A and B), wherein 10 ng ρTLPC6 plasmid-DNA (Fig. 1, SEQ ID NO: 2) was employed as template together with 50 pmol of each pair of primers (SEQ ID NO: 3 and SEQ ID NO: 4 or SEQ ID NO: 5 and SEQ ID NO: 6, respectively), which were synthesized according to the conventional phosphoramidite method. In addition, the reaction mixture contained 10 μl 10 x Taq buffer (100 mM Tris/HCl pH 9.0, 500 mM KC1, 15 mM MgCl2, 1% v/v Triton X-100) and 2 μl dNTP-Mix (10 mM dATP, dCTP, dGTP, dTTP). After bringing to volume with water, 5 U Taq DNA-polymerase (5 U/μl, Promega) were added and 20 cycles of 1 minute at 94°C, 1 minute at 62°C and 1.5 minutes at 72°C were carried out in a thermocycler with a heated lid (Eppendorf), followed by an incubation for 5 minutes at 60°C for final extension. The desired amplification products were isolated by preparative agarose gel electrophoresis from GTQ Agarose (Roth) using the Jetsorb DNA extraction kit (Genomed).
For the subsequent amplification step a 2000 μl mixture was prepared, wherein approximately 1000 fmol of both of these respective fragments were used as templates, in the presence of 1000 pmol of each of the assembly primers SEQ ID NO: 7, SEQ ID NO: 8 and 20 pmol of the mediating primer SEQ ID NO: 9. Both assembly primers had a biotin group at their 5'-ends allowing the purification of the PCR-product after Z?,stXI cleavage via streptavidin-coated paramagnetic beads. Additionally, 200 μl 10 x Taq buffer, 40 μl dNTP-Mix (10 mM dATP, dCTP, dGTP, dTTP), 100 u Taq DNA-polymerase (5 U/μl, Promega) and water were added to bring the mixture to the final volume of 2000 μl. The mixture was divided into 100 μl aliquots and PCR was performed with 20 cycles of 1 minute at 94°C, 1 minute at 60°C, 1.5 minutes at 72°C, followed by a subsequent incubation for 5 minutes at 60°C. The PCR product was purified using the E.Z.N.A. Cycle-Pure Kit (PeqLab).
For cloning purposes, this fragment representing the library of TPLC muteins in nucleic acid form was first cut with the restriction enzyme .BstXI (Promega) according to the instructions of the manufacturer and then purified by preparative agarose gel electrophoresis as described above, resulting in a double stranded DNA-fragment of 303 nucleotides in size. DNA-fragments not or incompletely digested were removed via their 5 '-biotin tags using streptavidin-coated paramagnetic beads (Merck). Therefore, 200 μl of the commercially available suspension of the paramagnetic particles in a concentration of 10 mg/ml were washed three times with 100 μl TE-buffer. The particles were then drained and mixed with 100 pmol of the DNA-fragment in 100 μl TE-buffer for 15 minutes at room temperature. The paramagnetic particles were collected at the wall of the Eppendorf vessel with the aid of a magnet and the supernatant containing the purified DNA fragment was recovered for further use in the following ligation reaction.
The DNA of the vector pTLPC7 (Fig.2) was cut with BstXI as described above and the larger of the two resulting fragments (3907 bp) was purified by preparative agarose gel electrophoresis.
For the ligation reaction, 5.99 μg (30 pmol) of the PCR fragment and 77.3 μg (30 pmol) of the vector fragment were incubated in the presence of 833 Weiss Units of T4 DNA ligase (Promega) in a total volume of 8330 μl (50 mMTris/HClpH 7.8, 10 mMMgCl2, lO mMDTT, l mMATP,
50 μg/ml BSA) for 24 h at 16°C. The DNA in the ligation mixture was then precipitated by adding 208 μl yeast tRNA (10 mg/ml solution in H O (Roche)), 8330 μl 5 M ammonium acetate, and 33.3 ml ethanol. Incubation at RT for 1 h was followed by centrifugation (30 minutes, 16000 g, 4°C). The precipitate was washed with 5 ml ethanol (70% v/v, RT), centrifuged (10 minutes,
16000 g, 4°C), and air dried until the DNA pellet appeared glossy and uncolored. Finally, the
DNA was dissolved to a final concentration of 200 μg/ml in a total volume of 416.5 μl water.
The preparation of electrocompetent E. coli XLl-Blue (Bullock et al., supra) was carried out according to the methods described by Tung and Chow (Trends Genet. 11 (1995), 128-129) and by Hengen (Trends Biochem. Sci. 21 (1996), 75-76). 1 1 LB-medium was adjusted to an optical density at 600 nm of OD60o = 0.08 by addition of a stationary XLl-Blue overnight culture and was incubated at 140 rpm and 26°C in a 21 Erlenmeyer flask. After reaching an OD6o0 = 0.6, the culture was cooled for 30 minutes on ice and subsequently centrifuged for 15 minutes at 4000 g and 4°C. The cells were washed twice with 500 ml ice-cold 10% w/v glycerol and finally re- suspended in 2 ml of ice-cold GYT-medium (10% w/v glycerol, 0.125% w/v yeast extract, 0.25% w/v tryptone). The cells were then aliquoted (200 μl), shock-frozen in liquid nitrogen and stored at -80°C.
The Micro Pulser system (BioRad) was used in conjunction with cuvettes from the same vendor (electrode separation 2 mm) for electroporation. All steps were carried out at room temperature employing pre-chilled cuvettes at a temperature of -20°C. Each 10 μl of the DNA solution (2 μg) was mixed with 100 μl of the cell suspension, incubated for 1 minute on ice, and transferred to the pre-chilled cuvette. Electroporation was performed (5 ms, 12.5 kV/cm) and the suspension was immediately diluted in 2 ml SOC-medium, followed by incubation for 60 minutes at 37°C and 140 rpm. Afterwards, the culture was diluted in 4 1 2 x YT-medium containing 100 μg/ml ampicillin (2 YT/Amp) resulting in an OD55o of 0.26. By employing a total of 78.61 μg ligated DNA about 1.0 x 1010 transformants were obtained in 42 electroporation runs.
Example 2: Selection and identification of TLPC muteins with determinable binding affinity towards a given target
The selection and identification of muteins of the invention with determinable binding affinity towards a given target can be carried in accordance with any protocol known to the skilled persons, for example, in accordance the respective protocols described in any of the PCT publications WO 99/16873, WO 00/75308, WO 03/029463 and WO 03/029471 as well the protocols described above. Principally, in a first step TLPC muteins can be presented and selected by means of phagemids to a given target that is immobilized on a solid phase. Then, selected muteins can be identified.
A more detailed suitable protocol for the generation of muteins that can bind to proteins is described in the following. As exemplary protein CD47 can be used.
Example 2.1: Production and purification of the extracellular domain of CD47
For the production of CD47 as a target protein for the isolation of CD47-specific TLPC muteins, E. coli JM83 (Yanisch-Perron et al. (1985) Gene 33, 103-119) was transformed with the expression plasmid pASK75-strepII-CD47 (Fig. 5) harboring the cDNA encoding the extracellular domain of CD47 (AS 1-119 of the mature protein) with a single mutation resulting in a Cys→Ala exchange at amino acid position 15 (for the CD47 cDNA, see Lindberg et al. (1993) J. Cell Biol. 123, 485-496). 100 ml LB-medium containing 100 μg/ml ampicillin (LB/Amp) was inoculated with a single colony of the JM83 transformant, and incubated overnight at 37°C, 160 rpm. Then, 2 1 LB/Amp-medium in a 5 1-Εrlenmeyer flask were inoculated with 40 ml of this pre-culture and were shaken at 26°C, 160 rpm to an OD550 = 0.4. Subsequently, the temperature was lowered to 22°C and upon reaching an OD550 = 0.5 the production of the recombinant protein was induced by adding 200 μg/1 anhydro-tetracycline (200 μl of a 2 mg/ml stock solution in DMF) followed by shaking for 3-4 further hours at 22°C and 160 rpm. The cells from one flask were centrifuged (10 minutes, 5500 g, 4°C) and re-suspended in 20 ml periplasmic release buffer (100 mM Tris/HCl pH 8.0, 500 mM sucrose, 1 mMEDTA), followed by cooling on ice for 30 minutes. Subsequently, the spheroplasts were removed in two successive centrifugation steps (25 minutes, 5300 g, 4°C and 10 minutes, 27000 g, 4°C, respectively). The supernatant containing the periplasmic protein extract was dialyzed against SA-buffer (100 mM Tris/HCl pH 8.0, 150 mM NaCl, 1 mM EDTA), sterile-filtered, and subjected to chromatographic purification.
The purification was performed by using the Strep-Tag® π-affinity tag (Skerra and Schmidt (2000) Methods Enzymol. 326, 271-304) located at the C-terminus of the CD47 protein and by employing Streptactin Superflow material (IB A). A chromatographic column with abed volume of 8 ml was filled with this affinity matrix and equilibrated with 20 ml SA-buffer at 4°C at a flow rate of 60 ml/h. Chromatography was monitored by measuring the absorption at 280 nm of the eluate in a flow-through photometer. After application of the periplasmic protein extract, the column was washed with SA-buffer until reaching the base line and the bound CD47 was subsequently eluted with ca. 15 ml of a solution of 2.5 mM D-desthiobiotin (IB A) in SA-buffer collecting fractions of the eluate. The fractions containing purified CD47 were analyzed via SDS- polyacrylamide gel electrophoresis (Fling und Gregerson (1986) Anal. Biochem. 155, 83-88), combined and dialyzed against an appropriate buffer, if needed. The protein yield was approximately 370 μg per 1 1 culture.
Example 2.2: Phagemid presentation and selection of TPLC muteins against the extracellular domain of human CD47 employing polystyrol sticks
A 2 ml aliquot of the stored phagemids from Example 1 containing a suitable number of phagemids (for example 1013 phagemids) can be centrifuged (30 minutes, 21460 g, 4 °C), the supernatant can then be removed, and the sedimented phagemid particles then dissolved in a buffer such as 1 ml PBS (4 mM KH2PO , 16 mM Na2HPO , 115 mM NaCl, pH 7.4). After incubation for 30 minutes on ice the solution is centrifuged (5 minutes, 18500 g, 4 °C) to remove residual aggregates.
Immuno-sticks (NUNC) can be used for the affinity enrichment of the recombinant phagemids carrying the BBP mutein fusion proteins. These sticks are first coated for a suitable period of time (for instance) overnight at 4 °C with 800 μl of the extracellular domain of human CD47 (CD47) (100 μg/ml) in SA-buffer as a target protein. Unoccupied binding sites on the surface of the Immuno-Stick are then saturated by incubation with 1.2 ml 2 % w/v BSA in PBST (PBS with 0.1 % v/v Tween 20) for 2 hours at room temperature. Afterwards, the Immuno-stick can be briefly washed for example three or four times with 1.2 ml PBST and subsequently incubated with a mixture of 500 μl of the phagemid solution (ca. 5»1012 cfu) and of 250 μl of PBS containing 6 % w/v BSA and 0.3 % v/v Tween 20 for 1 hour at room temperature.
For the removal of unbound phagemids, repeated washing can be performed, for examples six to eight times, each time with 950 μl PBST for 2 minutes. Adsorbed phagemids are then finally eluted by treating the Immuno-stick for 10 minutes with 950 μl 0.1 M glycine/HCl pH 2.2, followed by immediate neutralisation of the pH of the elution fraction by mixing it with 150 μl 0.5 M Tris.
For the amplification, this phagemid solution (1.1 ml, containing between 10 and 10 cfu, depending on the selection cycle) can be shortly warmed to 37 °C, mixed with 3 ml of an exponentially growing culture of E. coli XLl-blue (OD550 = 0.5), and incubated for 30 minutes at 37 °C, 140 rpm. The cells infected with the phagemids are subsequently sedimented (2 minutes, 4420 g, 4 °C), resuspended in 600 μl of the culture medium, and plated out onto three agar plates with LB/Amp-medium (LB/Amp agar; 145 mm diameter).
After incubation for 14 hours at 32 °C, the cells are scraped from the agar plates, each with addition of 10 ml 2xYT/Amp, are transferred to a sterile Erlenmeyer-flask, and are shaken for 30 minutes at 37 °C, 140 rpm for complete suspension. For the repeated production and affinity enrichment of phagemid particles 50 ml of 2xYT/Amp can be inoculated to an OD550 of ca. 0.08 with an appropriate volume of this suspension and incubated at 37 °C, 160 rpm until OD550 reached 0.5.
After infection with VCS-M13 helper phage (Stratagene) at a moi of approximately 10 the culture can be shaken for additional 45 minutes at 37 °C, 140 rpm. Kanamycin (70 μg/ml) is subsequently added, the incubator temperature lowered to 26 °C and, after 10 minutes, anhydrotetracycline is added at 25 μg/1 (6.2 μl of a 0.2 mg/ml stock solution in DMF) in order to induce gene expression. Incubation can continue for another 12-15 hours at 26 °C, 140 rpm.
The cells are sedimented by centrifugation (15 minutes, 12100 g, 4 °C) and the supernatant containing the phagemid particles sterile-filtered (0.45 μm), mixed with 1/4 volume (12.5 ml) 20 % w/v PEG 8000, 15 % w/v NaCl, and incubated on ice for 1 hour. After centrifugation (20 minutes, 18000 g, 4 °C) the precipitated phagemid particles are dissolved in 2 ml of cold BBS/E. The solution can be distributed to two 1.5 ml reaction vessels and incubated on ice for 30 minutes. After centrifugation of undissolved components (5 minutes, 21460 g, 4 °C) each supernatant is transferred to a new reaction vessel.
Mixture with 1/4 volume 20 % w/v PEG 8000, 15 % w/v NaCl and incubation for 30 to 60 minutes on ice can serve to reprecipitate the phagemid particles. After centrifugation (20 minutes, 21460 g, 4 °C) the supernatant is removed and the precipitated phagemid particles are dissolved and combined in a total of 1 ml PBS. After incubation for 30 minutes on ice the solution is centrifuged (5 minutes, 21460 g, 4 °C) in order to remove residual aggregates and the complete supernatant containing between 10π and 5»1012 phagemids is directly used for the next round of affinity enrichment. An appropriate number of further selection cycles (usually 4 or 5) with the CD47 can be carried out in this way.
Example 2.3: Identification of CD47-binding BBP muteins by use of a high- throughput ELISA screening method
For the analytical production of the TLPC muteins equipped with an N-terminal T7 detection tag (Novagen) as well as a C-terminal Strep-tag® II affinity tag and their characterization by high- throughput ELISA screening, a vector analogues to the vector the pBBP46 (Fig.7) can be used. The gene cassette containing the TLPC between the two BstXI cleavage sites can be subcloned from the vector pTLPC7 into the vector analogues to pBBP46 (Fig. 7).
For this purpose the plasmid DNA is isolated from the mixture of the E. coli clones obtained by infection with the phagemids from Example 2.2 eluted as a result of the last selection cycle, using the Plasmid Miniprep Spin kit (Genomed). The DNA is cut with the restriction enzyme BstXI and the smaller of the two fragments is purified by preparative agarose-gel electrophoresis as described in Example 1. The DNA of the vector that is analogues to pBBP46 is likewise cut with BstXI and the larger of the two fragments (4165 bp) was isolated in the same way.
For the ligation, each 50 fmol of the two DNA-fragments are mixed with 3 Weiss Units T4 DNA ligase (Promega) in a total volume of 20 μl (30 mM Tris/HCl pH 7.8, 10 mM MgCl2, 10 mM DTT, 1 mM ATP), followed by incubation for 2 h at 22 °C. E. coli TG1-F (E. coli K12 TGI, which had lost its episome) is transformed with 5 μl of this ligation mixture according to the CaCl2-method (Sambrook et al., supra) and plated on LB/ Amp agar plates (22 cm x 22 cm).
Single E. coli colonies obtained after the transformation harbouring the respective TLPC plasmids coding for the TLPC muteins are picked from these agar plates into 70 μl per well 2xYT/Amp in flat bottom 384 well plates (Greiner) by means of an automated colony picker (Genetix) and grown overnight at 37 °C at 700 rpm on a benchtop shaker (Bilhler) in a humidified incubator (MMM Medcenter) at 60 % relative humidity (rH). The cultures can be diluted 1:100 into 100 μl 2xYT/Amp in round bottom 96 well plates (Nunc) by means of a 96 pin replicating head (Genetix) and grown for ca. 1 h at 37 °C and 60 % rH, followed by an incubation for 3 h at 22 °C and 60 % rH, both at 700 rpm, until the OD55o reached approximately 0.6. The 384 well plates are kept as "master" plates at -80 °C after adding 25 μl 60 % v/v glycerol to each well.
Recombinant TLPC muteins are produced in the 96 well plates by adding 20 μl per well of 1.2 μg/ml anhydrotetracyclin in 2xYT (obtained by diluting a 2 mg/ml stock solution 1:1667 in 2xYT; final concentration 0.2 μg/ml) to the bacterial cultures and incubation overnight at 22 °C and 700 rpm at 60 % rH. Afterwards, 40 μl of lysis buffer (400 mM Na-borate pH 8.0, 320 mM NaCl, 4 mM EDTA, 0.3 % w/v lysozyme) is added to each well and the plate was incubated for 1 h at 22 °C and 700 rpm at 60 % rH. To minimize non-specific binding interactions in the subsequent ELISA experiment, obtained crude cell extracts can be supplemented with 40 μl/well PBS containing 10 % w/v BSA and 0.05 % v/v Tween 20 (final concentration 2 % BSA) for 1 h at 22 °C and 700 rpm at 60 % rH.
For the detection of binding, the crude cell extracts containing the TLPC muteins are tested for their reactivity with the prescribed target protein CD47 and the unrelated control protein aldolase
(Roche), respectively, in ELISA experiments. Therefore, wells of black Fluotrac 600 ELISA plates (Greiner; 384 well) are coated overnight with 20 μl of a solution of recombinant CD47 (produced as described in Example 2.1) or the control protein at 4 °C, each at a concentration of 20 μg/ml in PBS. Plates are washed five times with 100 μl PBS containing 0.05 % v/v Tween 20 (PBST/0.05) per well with an automated ELISA plate washer (Molecular Devices) leaving a residual volume of 10 μl of the washing buffer in each well after the last washing step. Residual binding sites are blocked by incubation with 100 μl PBST/0.05 containing 2 % w/v BSA for 2 h at room temperature. Afterwards, plates are again washed five times as described above.
For complex formation between the TLPC muteins and the immobilized proteins, the wells can be incubated with 10 μl of the cell extract from above for 1 hour at room temperature. Subsequently, plates are washed again five times and 10 μl of an anti-T7 monoclonal antibody- HRP-conjugate (Amersham), diluted 1 :5000 in PBST/0.05 containing 0.5 % w/v non-fat dry milk powder (Vitalia), is added to each well and incubated for 1 hour at room temperature. Plates are again washed five times and 10 μl of the fluorogenic HRP-substrate QuantaBlu™ (Pierce, diluted as described by the manufacturer) is added to detect bound TPLC muteins by means of the attached anti-T7 monoclonal antibody-HRP-conjugate. After 60 minutes at room temperature fluorescence is excited at a wavelength of 320 nm (± 12.5 nm) and measured at 430 nm (± 17.5 nm) in a GENiosPlus plate reader (Tecan).
Those analyzed TLPC muteins that showed a binding signal on the prescribed target protein (CD47) which exceeded the binding signal on the unrelated control protein (aldolase) at least by a factor of three (signal-to-control ratio of at least 3) can be subsequently subjected to a secondary high-throughput ELISA screening experiment in order to study the binding specificity of these muteins in greater detail. Therefore, these clones are transferred from the 384 well master plates described above onto LB/Amp agar, and grown overnight at 37 °C. 100 μl 2xYT/Amρ in round bottom 96 well plates (Nunc) was inoculated with single colonies from these agar plates and grown overnight at 37 °C at 700 rpm and 60 % rH. The cultures are diluted 1:100 into 100 μl 2xYT/Amp in round bottom 96 well plates (Nunc) and production of recombinant TLPC muteins as well as preparation of the bacterial lysates was performed as described above.
For the detection of target-specificity of the TLPC muteins, wells of black Fluotrac 600 ELISA plates (Greiner; 384 well) are coated overnight at 4 °C with 20 μl of a solution of recombinant CD47 or, as a control, with BSA (Roth), transferrin (Roche), aldolase (Roche), ovalbumin (Sigma) as well as a conjugate of RNaseA (Fluka) and digoxigenin, each at a concentration of 20 μg/ml in PBS.
This conjugate is prepared by reacting RNaseA at a twofold molar ratio of digoxigenin-3-O- methylcarbonyl-ε-amidocaproic acid-N-hydroxy-succinimide ester (DIG-NHS; Roche) according to the instructions of the manufacturer. Excess reactant is removed from the RNaseA-conjugate by means of size exclusion chromatography using a HiTrap column (Amersham) according to the instructions of the manufacturer employing PBS as running buffer.
After overnight incubation, the plates are washed as described above and blocked by the addition of 100 μl/well PBST/0.05 containing 2 % w/v BSA at the conditions described above, followed again by washing of the plates. 10 μl of the blocked bacterial lysates of the selected TLPC muteins mentioned above are transferred to each of the wells coated with either CD47 or the unrelated control proteins and incubated for 1 h at ambient temperature. Bound TPLC muteins are detected with anti-T7 monoclonal antibody-HRP-conjugate and the fluorogenic HRP-substrate QuantaBlu™ as described above.
The nucleotide sequence of chosen TLPC clones can then be determined. Furthermore, these clones can then be chosen for the determination of their binding affinities for CD47.

Claims

What is claimed is:
1. A mutein derived from a polypeptide of tear lipocalin or a functional homologue thereof, wherein the mutein comprises at least two mutated amino acid residues at any sequence position in the N-terminal peptide stretch as well as in the three peptide loops BC, DE, and FG arranged at the end of the β-barrel structure that is located opposite of the natural lipocalin binding pocket, wherein said tear lipocalin or functional homologue thereof has at least 60% sequence homology with human tear lipocalin, and wherein the mutein binds a given target with detectable affinity.
2. The mutein of claim 1, wherein the mutein is derived from human tear lipocalin.
3. The mutein of claim 1 or 2, wherein the mutein comprises amino acid mutations at any two or more of the sequence positions in the peptide segments corresponding to the sequence positions 7-14, 43-49, 70-77, and 87-97 of the linear polypeptide sequence of human tear lipocalin.
4. The mutein of any of claims 1 to 3, wherein the mutein comprises amino acid mutations at any two or more of the sequence positions corresponding to the sequence positions 8, 9, 10, 11, 12, 13, 43, 45, 47, 70, 72, 74, 75, 90, 92, 94, and 97 of the linear polypeptide sequence of human tear lipocalin.
5. A mutein derived from a polypeptide of tear lipocalin or a functional homologue thereof, wherein the mutein comprises at least two mutated amino acid residues at any sequence position in the four peptide loops AB, CD, EF, and GH encompassing the natural lipocalin binding pocket, wherein said tear lipocalin or functional homologue thereof has at least 60% sequence homology with human tear lipocalin, and wherein the mutein binds a given target with detectable affinity.
6. The mutein of claim 5, wherein the mutein is derived from human tear lipocalin.
7. The mutein of claim 5 or 6, wherein the mutein comprises amino acid mutations at any two or more of the sequence positions in the peptide segments corresponding to the sequence positions 24-36, 53-66, 79-84, and 103-110 of the linear polypeptide sequence of human tear lipocalin.
8. The mutein of any of claims 5 to 7, further comprising at least two mutated amino acid residues at any sequence position in the N-terminal peptide stretch as well as the three peptide loops BC, DE, and FG arranged at the end of the β-barrel structure that is located opposite of the natural lipocalin binding pocket.
9. The mutein of claim 8, comprising amino acid mutations at any two or more of the sequence positions in the peptide segments corresponding to the sequence positions 7-14, 43-49, 70-77, and 87-97 of the linear polypeptide sequence of human tear lipocalin.
10. The mutein of claim 8 or 9, comprising amino acid mutations at any two or more of the sequence positions corresponding to the sequence positions 8, 9, 10, 11, 12, 13, 43, 45, 47, 70, 72, 74, 75, 90, 92, 94, and 97 of the linear polypeptide sequence of human tear lipocalin.
11. A mutein derived from a polypeptide of tear lipocalin or a functional homologue thereof, wherein the mutein comprises at least two mutated amino acid residues at any sequence position in the N-terminal region as well as the three peptide loops BC, DE, and FG arranged at the end of the β-barrel structure that is located opposite of the natural lipocalin binding pocket, wherein the mutein comprises at least two mutated amino acid residues at any sequence position in the four peptide loops AB, CD, EF, and GH encompassing the natural lipocalin binding pocket, wherein said tear lipocalin or functional homologue thereof has at least 60% sequence homology with human tear lipocalin, and wherein the mutein binds at least one given target with detectable affinity.
12. The mutein of any of claims 1 to 10, wherein the mutein is conjugated to a label selected from the group consisting of organic molecules, enzyme labels, radioactive labels, colored labels, fluorescent labels, chromogenic labels, luminescent labels, haptens, digoxigenin, biotin, metal complexes, metals, and colloidal gold.
13. The mutein of any of claims 1 to 13, wherein the mutein is fused at its N-terminus or its C- terminus to a protein, a protein domain or a peptide.
14. A nucleic acid molecule comprising a nucleotide sequence encoding a mutein of any of claims 1 to 13.
15. The nucleic acid molecule of claim 15 comprised in a vector.
16. The nucleic acid molecule of claim 15 comprised in a phagemid vector.
17. A host cell containing a nucleic acid molecule of any of claims 14 to 16.
18. A method for the generation of a mutein of any of claims 1 to 13, comprising: (a) subjecting a nucleic acid molecule encoding a tear lipocalin of a functional homologue thereof, wherein said tear lipocalin or functional homologue thereof has at least 60% sequence homology with human tear lipocalin, to mutagenesis at two or more different nucleotide triplets, (b) expressing the at least one mutein nucleic acid molecules obtained in (a) in a suitable expression system, and (c) enriching at least one mutein having a detectable binding affinity for a given target by means of selection and/or isolation.
19. A method for the production of a mutein according to any of claims 1 to 13, wherein the mutein, a fragment of the mutein or a fusion protein of the mutein and another polypeptide is produced starting from the nucleic acid coding for the mutein by means of genetic engineering methods.
20. The method of claim 19, wherein the mutein is produced in a bacterial or eukaryotic host organism and is isolated from this host organism or its culture.
21. A pharmaceutical composition comprising at least one mutein of any of claims 1 tol3.
22. Use of a mutein of any of claims 1 to 13 for the detection of a given target, comprising the steps of: (a) contacting the mutein with a test sample supposed to contain said target, and (b) detecting of the mutein/target complex by a suitable signal.
23. Use of claim 22, wherein the given target is a protein or protein domain, a peptide, a nucleic acid molecule, an organic molecule or a metal complex and the detection is carried out for validation of the protein as pharmacological drug target.
24. Use of a mutein of any of claims 1 to 13 for the separation of a given target, comprising: (a) contacting the mutein with a sample supposed to contain said target, and (b) separating the mutein/target complex from the sample.
25. The use of any of claim 22 to 24, wherein the mutein/target complex is bound onto a solid phase.
26. Use of a mutein of any of claims 1 to 13 for the targeting of a compound to a preselected site comprising (a) contacting the mutein with said compound, and (b) delivering the mutein/compound complex to the preselected site
27. Use of a mutein of any of claims 1 to 13 for complex formation with a given target.
PCT/EP2003/009404 2003-08-25 2003-08-25 Muteins of tear lipocalin WO2005019255A1 (en)

Priority Applications (16)

Application Number Priority Date Filing Date Title
PCT/EP2003/009404 WO2005019255A1 (en) 2003-08-25 2003-08-25 Muteins of tear lipocalin
AU2003275958A AU2003275958A1 (en) 2003-08-25 2003-08-25 Muteins of tear lipocalin
DK04764425T DK1660528T3 (en) 2003-08-25 2004-08-24 Mutein of tear-lipocalin
AT04764425T ATE446313T1 (en) 2003-08-25 2004-08-24 MUTEINE OF TEAR LIPOCALIN
JP2006524319A JP4856543B2 (en) 2003-08-25 2004-08-24 Tear lipocalin mutein
BRPI0413728A BRPI0413728B8 (en) 2003-08-25 2004-08-24 human tear lipocalin mutein, methods for generating and producing mutein, pharmaceutical composition and use of mutein
CA2536491A CA2536491C (en) 2003-08-25 2004-08-24 Muteins of tear lipocalin
SG200701410-3A SG131109A1 (en) 2003-08-25 2004-08-24 Muteins of tear lipocalin
PCT/EP2004/009447 WO2005019256A2 (en) 2003-08-25 2004-08-24 Muteins of tear lipocalin
AU2004266846A AU2004266846B2 (en) 2003-08-25 2004-08-24 Muteins of tear lipocalin
US10/569,134 US7585940B2 (en) 2003-08-25 2004-08-24 Muteins of human tear lipocalin
CN2004800313369A CN1871256B (en) 2003-08-25 2004-08-24 Muteins of tear lipocalin
DE602004023725T DE602004023725D1 (en) 2003-08-25 2004-08-24 MUTINES OF TIN-LIPOCALIN
EP04764425A EP1660528B1 (en) 2003-08-25 2004-08-24 Muteins of tear lipocalin
HK06106847.6A HK1084404A1 (en) 2003-08-25 2006-06-14 Muteins of tear lipocalin
US12/461,633 US7893208B2 (en) 2003-08-25 2009-08-19 Muteins of human tear lipocalin

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
PCT/EP2003/009404 WO2005019255A1 (en) 2003-08-25 2003-08-25 Muteins of tear lipocalin

Publications (2)

Publication Number Publication Date
WO2005019255A1 true WO2005019255A1 (en) 2005-03-03
WO2005019255A8 WO2005019255A8 (en) 2005-05-26

Family

ID=34203142

Family Applications (2)

Application Number Title Priority Date Filing Date
PCT/EP2003/009404 WO2005019255A1 (en) 2003-08-25 2003-08-25 Muteins of tear lipocalin
PCT/EP2004/009447 WO2005019256A2 (en) 2003-08-25 2004-08-24 Muteins of tear lipocalin

Family Applications After (1)

Application Number Title Priority Date Filing Date
PCT/EP2004/009447 WO2005019256A2 (en) 2003-08-25 2004-08-24 Muteins of tear lipocalin

Country Status (13)

Country Link
US (2) US7585940B2 (en)
EP (1) EP1660528B1 (en)
JP (1) JP4856543B2 (en)
CN (1) CN1871256B (en)
AT (1) ATE446313T1 (en)
AU (2) AU2003275958A1 (en)
BR (1) BRPI0413728B8 (en)
CA (1) CA2536491C (en)
DE (1) DE602004023725D1 (en)
DK (1) DK1660528T3 (en)
HK (1) HK1084404A1 (en)
SG (1) SG131109A1 (en)
WO (2) WO2005019255A1 (en)

Cited By (45)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008015239A2 (en) 2006-08-01 2008-02-07 Pieris Ag Muteins of tear lipocalin and methods for obtaining the same
WO2008063136A1 (en) 2006-11-24 2008-05-29 Agency For Science, Technology And Research Apparatus for processing a sample in a liquid droplet and method of using the same
US7611847B2 (en) 2005-06-01 2009-11-03 Agency For Science, Technology And Research Method for identifying an intestinal phenotype
EP2189539A1 (en) 2008-11-21 2010-05-26 Chimera Biotec GmbH Conjugate complexes for analyte detection
WO2011015634A2 (en) 2009-08-05 2011-02-10 Pieris Ag Controlled release formulations of lipocalin muteins
WO2011069992A2 (en) 2009-12-07 2011-06-16 Pieris Ag Muteins of human lipocalin 2 (lcn2, hngal) with affinity for a given target
WO2011078794A1 (en) 2009-12-22 2011-06-30 Agency For Science, Technology And Research Sers-based analyte detection
WO2011154420A2 (en) 2010-06-08 2011-12-15 Pieris Ag Tear lipocalin muteins binding il-4 r alpha
WO2012022742A1 (en) 2010-08-16 2012-02-23 Pieris Ag Binding proteins for hepcidin
WO2012065978A1 (en) 2010-11-15 2012-05-24 Pieris Ag Muteins of human lipocalin 2 with affinity for glypican-3 (gpc3)
WO2012072806A1 (en) 2010-12-02 2012-06-07 Pieris Ag Muteins of human lipocalin 2 with affinity for ctla-4
US8216855B2 (en) 2006-02-13 2012-07-10 Agency For Science, Technology And Research Method of processing a biological and/or chemical sample
US8261598B2 (en) 2006-03-09 2012-09-11 Agency For Science, Technology And Research Apparatus for performing a reaction in a droplet and method of using the same
WO2013174783A1 (en) 2012-05-23 2013-11-28 Pieris Ag Lipocalin muteins with binding-affinity for glypican-3 (gpc-3) and use of lipocalin muteins for target-specific delivery to cells expressing gpc-3
WO2013189521A1 (en) 2012-06-19 2013-12-27 Waclawczyk Simon Method of generating cells of hepatocyte phenotype
US8663915B2 (en) 2007-04-13 2014-03-04 Agency For Science, Technology And Research Methods of controlling tumorigenesis and diagnosing the risk thereof
US8716420B2 (en) 2008-10-13 2014-05-06 Agency For Science, Technology And Research Amphiphilic polymers and nanocrystals coated therewith
US8784752B2 (en) 2009-04-17 2014-07-22 Curiox Biosystems Pte Ltd Use of chemically patterned substrate for liquid handling, chemical and biological reactions
WO2014140210A1 (en) 2013-03-14 2014-09-18 Pieris Ag Novel binding proteins for pcsk9
US8871686B2 (en) 2009-07-07 2014-10-28 Agency For Science, Technology And Research Methods of identifying a pair of binding partners
US8969313B2 (en) 2007-11-02 2015-03-03 Agency For Science, Technology And Research Methods and compounds for preventing and treating a tumour
EP2899277A1 (en) 2004-11-26 2015-07-29 Pieris AG Compound with affinity for the cytotoxic T lymphocyte-associated antigen (CTLA-4)
US9102920B2 (en) 2008-05-06 2015-08-11 Agency For Science, Technology And Research Method of effecting de-differentiation of a cell
WO2015140314A1 (en) 2014-03-21 2015-09-24 Philipps-Universität Marburg Means and methods for itaconic acid production
WO2016142314A1 (en) 2015-03-06 2016-09-15 Deutsches Krebsforschungszentrum FUSION PROTEINS COMPRISING A BINDING PROTEIN AND AN INTERLEUKIN-15 POLYPEPTIDE HAVING A REDUCED AFFINITY FOR IL15Ra AND THERAPEUTIC USES THEREOF
WO2016184875A1 (en) 2015-05-18 2016-11-24 Pieris Pharmaceuticals Gmbh Muteins of human lipocalin 2 with affinity for glypican-3 (gpc3)
US9557318B2 (en) 2013-07-09 2017-01-31 Curiox Biosystems Pte Ltd. Array plates for washing samples
US9874501B2 (en) 2006-11-24 2018-01-23 Curiox Biosystems Pte Ltd. Use of chemically patterned substrate for liquid handling, chemical and biological reactions
US9878328B2 (en) 2010-07-23 2018-01-30 Curiox Biosystems Pte Ltd. Apparatus and method for multiple reactions in small volumes
US9950323B2 (en) 2012-02-05 2018-04-24 Curiox Biosystems Pte Ltd. Array plates and methods for making and using same
EP3381933A1 (en) 2008-06-24 2018-10-03 Technische Universität München Muteins of hngal and related proteins with affinity for a given target
US10400016B2 (en) 2015-12-10 2019-09-03 Pieris Pharmaceuticals Gmbh Proteins specific for calcitonin gene-related peptide
US10398754B2 (en) 2011-12-13 2019-09-03 Pieris Pharmaceuticals Gmbh Methods for preventing or treating certain disorders by inhibiting binding of IL-4 and/or IL-13 to their respective receptors
WO2019243564A1 (en) 2018-06-21 2019-12-26 Technische Universitaet Muenchen Hepatitis b and/or hepatitis d-permissive cells and animals
US10526384B2 (en) 2012-11-19 2020-01-07 Pieris Pharmaceuticals Gmbh Interleukin-17A-specific and interleukin-23-specific binding polypeptides and uses thereof
US10526382B2 (en) 2015-01-28 2020-01-07 Pieris Pharmaceuticals Gmbh Human neutrophil gelatinase-associated lipocalin (hNGAL) muteins capable of binding angiopoietin-2 (Ang-2) and methods of use thereof
US10545139B2 (en) 2015-06-16 2020-01-28 Curiox Biosystems Pte Ltd. Methods and devices for performing biological assays using magnetic components
US10703810B2 (en) 2015-11-30 2020-07-07 Pieris Australia Pty Ltd. Fusion polypeptides which bind vascular endothelial growth factor a (VEGF-A) and angiopoietin-2 (Ang-2)
US10725020B2 (en) 2007-11-14 2020-07-28 Curiox Biosystems Pte Ltd. High throughput miniaturized assay system and methods
US10774119B2 (en) 2014-05-22 2020-09-15 Pieris Pharmaceuticals Gmbh Specific-binding polypeptides and uses thereof
US10865250B2 (en) 2015-05-04 2020-12-15 Pieris Pharmaceuticals Gmbh Anti-cancer fusion polypeptide
US10913778B2 (en) 2015-05-18 2021-02-09 Pieris Pharmaceuticals Gmbh Anti-cancer fusion polypeptides, encoding nucleic acids and methods of using polypeptides
US10927154B2 (en) 2014-01-13 2021-02-23 Pieris Pharmaceuticals Gmbh Multi-specific polypeptide useful for localized tumor immunomodulation
US11261221B2 (en) 2015-05-04 2022-03-01 Pieris Pharmaceuticals Gmbh Proteins specific for CD137
US11692162B2 (en) 2017-04-05 2023-07-04 Curiox Biosystems Pte Ltd. Methods, devices, and apparatus for washing samples on array plates

Families Citing this family (123)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU2003275958A1 (en) * 2003-08-25 2005-03-10 Pieris Proteolab Ag Muteins of tear lipocalin
WO2007107563A2 (en) * 2006-03-20 2007-09-27 Technische Universität München Muteins of tear lipocalin with affinity for the t-cell coreceptor cd4
CN101516907B (en) * 2006-08-01 2015-08-26 皮里斯股份公司 The mutain of tear lipocalin and preparation method thereof
US20090148905A1 (en) 2007-11-30 2009-06-11 Claire Ashman Antigen-binding constructs
US9212208B2 (en) * 2008-01-30 2015-12-15 Pieris Ag Muteins with tear lipocalin having affinity to human c-Met receptor tyrosine kinase and methods for obtaining the same
US20090274623A1 (en) * 2008-01-30 2009-11-05 General Electric Company In vivo imaging agents for met receptor tyrosine kinase
US9051568B2 (en) 2008-04-25 2015-06-09 Rutgers, The State University Of New Jersey Anti-sense microrna expression vectors
EP2271323A2 (en) 2008-05-06 2011-01-12 Glaxo Group Limited Encapsulation of biologically active agents
JP2010074915A (en) * 2008-09-17 2010-04-02 Jtekt Corp Motor controller and electric power steering device
UY32341A (en) 2008-12-19 2010-07-30 Glaxo Group Ltd NEW ANTIGEN UNION PROTEINS
EP2401298A1 (en) 2009-02-24 2012-01-04 Glaxo Group Limited Antigen-binding constructs
CA2753332A1 (en) 2009-02-24 2010-09-02 Glaxo Group Limited Antigen-binding constructs
WO2010097394A1 (en) 2009-02-24 2010-09-02 Glaxo Group Limited Multivalent and/or multispecific rankl-binding constructs
EP2459053A2 (en) 2009-07-28 2012-06-06 F. Hoffmann-La Roche AG Non-invasive in vivo optical imaging method
SG182590A1 (en) 2010-01-28 2012-08-30 Glaxo Group Ltd Cd127 binding proteins
KR101341640B1 (en) * 2010-02-05 2013-12-16 한국전자통신연구원 The analying method of protein microstructure and array for the same
US20150231215A1 (en) 2012-06-22 2015-08-20 Randolph J. Noelle VISTA Antagonist and Methods of Use
NZ602634A (en) 2010-03-26 2015-06-26 Dartmouth College Vista regulatory t cell mediator protein, vista binding agents and use thereof
US10745467B2 (en) 2010-03-26 2020-08-18 The Trustees Of Dartmouth College VISTA-Ig for treatment of autoimmune, allergic and inflammatory disorders
MX2012012736A (en) 2010-05-07 2013-02-26 Hoffmann La Roche Diagnostic method for the detection of cells ex vivo.
AR081556A1 (en) 2010-06-03 2012-10-03 Glaxo Group Ltd HUMANIZED ANTIGEN UNION PROTEINS
US8987415B2 (en) 2010-09-07 2015-03-24 Technische Universitaet Muenchen Recombinant bacterial lipocalin blc and uses thereof
CN103201293B (en) * 2010-09-08 2016-04-27 哈洛齐梅公司 The method of assessment and qualification or development condition active therapeutic protein
KR101947356B1 (en) 2010-11-23 2019-02-12 글락소 그룹 리미티드 Antigen binding proteins to oncostatin m (osm)
MX2013005847A (en) 2010-11-24 2013-12-12 Glaxo Group Ltd Multispecific antigen binding proteins targeting hgf.
WO2012136685A1 (en) 2011-04-04 2012-10-11 Pieris Ag Methods and compositions for anti-vegf and anti-c-met therapy
CN103596974B (en) 2011-04-15 2016-08-31 卡姆普根有限公司 Polypeptide and polynucleotide and for treating immunity related disorder and the purposes of cancer
LT3415531T (en) 2011-05-27 2023-09-25 Glaxo Group Limited Bcma (cd269/tnfrsf17) - binding proteins
WO2013048986A1 (en) 2011-09-26 2013-04-04 Knoa Software, Inc. Method, system and program product for allocation and/or prioritization of electronic resources
AU2012327878A1 (en) 2011-10-28 2014-05-29 Patrys Limited PAT-LM1 epitopes and methods for using same
SG11201402996WA (en) 2011-12-12 2014-07-30 Pieris Ag Methods for preventing or treating disorders by increasing bioavailability of iron and related pharmaceutical formulation
DK2797622T3 (en) 2011-12-30 2017-01-16 Halozyme Inc PH20 polypeptide variants, formulations and uses thereof
EP2802341B1 (en) 2012-01-09 2019-03-06 Pieris Pharmaceuticals GmbH Methods for preventing, treating or diagnosing disorders
EP2809679A1 (en) 2012-01-31 2014-12-10 Technische Universität München Muteins of a1m lipocalin and method of production therefor
HUE049860T2 (en) 2012-02-13 2020-11-30 Agency Science Tech & Res IL-ß NEUTRALIZING HUMAN MONOCLONAL ANTIBODIES
GB2502127A (en) 2012-05-17 2013-11-20 Kymab Ltd Multivalent antibodies and in vivo methods for their production
US9890215B2 (en) 2012-06-22 2018-02-13 King's College London Vista modulators for diagnosis and treatment of cancer
EP2892558B1 (en) 2012-09-07 2019-04-10 The Trustees Of Dartmouth College Vista modulators for diagnosis and treatment of cancer
SG11201502538TA (en) 2012-11-08 2015-05-28 Hoffmann La Roche Her3 antigen binding proteins binding to the beta-hairpin of her3
TW201534726A (en) 2013-07-03 2015-09-16 Halozyme Inc Thermally stable PH20 hyaluronidase variants and uses thereof
CA2926215A1 (en) 2013-10-06 2015-04-09 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Modified pseudomonas exotoxin a
TR201900665T4 (en) 2013-11-28 2019-02-21 B Creative Sweden Ab Treatment method of diabetic nephropathy.
KR20160096194A (en) 2013-12-18 2016-08-12 씨에스엘 리미티드 Method of treating wounds
US11014987B2 (en) 2013-12-24 2021-05-25 Janssen Pharmaceutics Nv Anti-vista antibodies and fragments, uses thereof, and methods of identifying same
JP6590810B2 (en) 2013-12-24 2019-10-16 ヤンセン ファーマシューティカ エヌブイ Anti-VISTA antibodies and fragments
MX2016016310A (en) 2014-06-11 2017-10-20 A Green Kathy Use of vista agonists and antagonists to suppress or enhance humoral immunity.
WO2016059602A2 (en) 2014-10-16 2016-04-21 Glaxo Group Limited Methods of treating cancer and related compositions
PL3224275T3 (en) 2014-11-14 2020-09-07 F.Hoffmann-La Roche Ag Antigen binding molecules comprising a tnf family ligand trimer
MX2017007136A (en) 2014-12-05 2017-12-04 Immunext Inc Identification of vsig8 as the putative vista receptor and its use thereof to produce vista/vsig8 modulators.
US11382963B2 (en) 2015-01-12 2022-07-12 Pieris Pharmaceuticals Gmbh Engineered T cells and uses therefor
JP7026509B2 (en) 2015-06-24 2022-02-28 ヤンセン ファーマシューティカ エヌブイ Anti-VISTA antibody and fragment
EP3115066A1 (en) 2015-07-07 2017-01-11 Technische Universität München Novel psma-specific binding proteins
MX2018000447A (en) 2015-07-15 2018-08-15 Pieris Pharmaceuticals Gmbh Novel proteins specific for lag-3.
WO2017025498A1 (en) 2015-08-07 2017-02-16 Pieris Pharmaceuticals Gmbh Novel fusion polypeptide specific for lag-3 and pd-1
AR106188A1 (en) 2015-10-01 2017-12-20 Hoffmann La Roche ANTI-CD19 HUMANIZED HUMAN ANTIBODIES AND METHODS OF USE
MA43017A (en) 2015-10-02 2018-08-08 Hoffmann La Roche BISPECIFIC ANTIBODIES SPECIFIC TO A TNF CO-STIMULATION RECEPTOR
LT3356411T (en) 2015-10-02 2021-09-10 F. Hoffmann-La Roche Ag Bispecific antibodies specific for pd1 and tim3
PE20181349A1 (en) 2015-10-07 2018-08-22 Hoffmann La Roche BIESPECIFIC ANTIBODIES WITH TETRAVALENCE FOR A CO-STIMULATING FNT RECEIVER
TW202216201A (en) 2016-02-12 2022-05-01 比利時商楊森製藥公司 Anti-vista antibodies and fragments, uses thereof, and methods of identifying same
EP3231813A1 (en) 2016-03-29 2017-10-18 F. Hoffmann-La Roche AG Trimeric costimulatory tnf family ligand-containing antigen binding molecules
RU2021111187A (en) 2016-04-15 2021-04-29 Янссен Фармасьютикалз, Инк. ANTIBODIES AGAINST HUMAN VISTA AND THEIR APPLICATION
CN109071652B (en) 2016-05-11 2022-09-23 豪夫迈·罗氏有限公司 Antigen binding molecules comprising TNF family ligand trimers and tenascin binding modules
EP3243832A1 (en) 2016-05-13 2017-11-15 F. Hoffmann-La Roche AG Antigen binding molecules comprising a tnf family ligand trimer and pd1 binding moiety
JP2020501508A (en) 2016-09-15 2020-01-23 クアドルセプト バイオ リミテッド Multimers, tetramers and octamers
JP7051826B2 (en) 2016-09-23 2022-04-11 シーエスエル、リミテッド Coagulation factor binding protein and its use
WO2018087108A1 (en) 2016-11-09 2018-05-17 Pieris Pharmaceuticals Gmbh Proteins specific for cd137
CA3039430A1 (en) 2016-12-19 2018-06-28 F. Hoffmann-La Roche Ag Combination therapy with targeted 4-1bb (cd137) agonists
KR20190093588A (en) 2016-12-20 2019-08-09 에프. 호프만-라 로슈 아게 Combination therapy of anti-CD20 / anti-CD3 bispecific antibodies and 4-1BB (CD137) agonists
BR112019013189A2 (en) 2017-01-03 2019-12-10 Hoffmann La Roche bispecific antigen binding molecules, polynucleotide, host cell, bispecific antigen binding molecule production method, pharmaceutical composition, use, methods for inhibiting tumor cell growth in an individual and for treating cancer or an infectious disease
US11168119B2 (en) * 2017-01-18 2021-11-09 Pieris Pharmaceuticals Gmbh Lipocalin muteins with binding affinity for LAG-3
WO2018134279A1 (en) 2017-01-18 2018-07-26 Pieris Pharmaceuticals Gmbh Novel fusion polypeptides specific for lag-3 and pd-1
WO2018178074A1 (en) 2017-03-29 2018-10-04 F. Hoffmann-La Roche Ag Trimeric antigen binding molecules specific for a costimulatory tnf receptor
EP3601346A1 (en) 2017-03-29 2020-02-05 H. Hoffnabb-La Roche Ag Bispecific antigen binding molecule for a costimulatory tnf receptor
EP3601345A1 (en) 2017-03-29 2020-02-05 H. Hoffnabb-La Roche Ag Bispecific antigen binding molecule for a costimulatory tnf receptor
MA49039A (en) 2017-04-04 2020-02-12 Hoffmann La Roche NEW BISPECIFIC ANTIGEN BINDING MOLECULES CAPABLE OF BINDING SPECIFICALLY TO CD40 AND FAP
SG11201909154SA (en) 2017-04-05 2019-10-30 Hoffmann La Roche Bispecific antibodies specifically binding to pd1 and lag3
KR20200067158A (en) 2017-10-18 2020-06-11 씨에스엘 리미티드 Human serum albumin variants and uses thereof
CN111246884A (en) 2017-11-01 2020-06-05 豪夫迈·罗氏有限公司 Novel antigen binding molecules comprising trimers of TNF family ligands
US20210324108A1 (en) 2017-11-01 2021-10-21 Hoffmann-La Roche Inc. Bispecific 2+1 contorsbodies
EP3704155A2 (en) 2017-11-01 2020-09-09 F. Hoffmann-La Roche AG Combination therapy with targeted ox40 agonists
ES2939461T3 (en) 2017-11-29 2023-04-24 Csl Ltd Method for treating or preventing ischemia-reperfusion injury
EP3502140A1 (en) 2017-12-21 2019-06-26 F. Hoffmann-La Roche AG Combination therapy of tumor targeted icos agonists with t-cell bispecific molecules
EP3765489A1 (en) 2018-03-13 2021-01-20 F. Hoffmann-La Roche AG Therapeutic combination of 4-1 bb agonists with anti-cd20 antibodies
AU2019235330A1 (en) 2018-03-13 2020-07-16 F. Hoffmann-La Roche Ag Combination therapy with targeted 4-1BB (CD137) agonists
AR114284A1 (en) 2018-04-13 2020-08-12 Hoffmann La Roche HER2 TARGETING ANTIGEN BINDING MOLECULES COMPOSING 4-1BBL
EP3818082A1 (en) 2018-07-04 2021-05-12 F. Hoffmann-La Roche AG Novel bispecific agonistic 4-1bb antigen binding molecules
TWI837156B (en) 2018-07-31 2024-04-01 德商皮里斯製藥有限公司 Novel fusion protein specific for cd137 and pd-l1
MX2021003548A (en) 2018-10-01 2021-05-27 Hoffmann La Roche Bispecific antigen binding molecules comprising anti-fap clone 212.
CN112654641A (en) 2018-10-01 2021-04-13 豪夫迈·罗氏有限公司 Bispecific antigen binding molecules with trivalent binding to CD40
CN113286821A (en) 2018-12-21 2021-08-20 豪夫迈·罗氏有限公司 Tumor-targeting agonistic CD28 antigen binding molecules
WO2020127628A1 (en) 2018-12-21 2020-06-25 F. Hoffmann-La Roche Ag Tumor-targeted superagonistic cd28 antigen binding molecules
MX2021010039A (en) 2019-02-26 2021-09-21 Pieris Pharmaceuticals Gmbh Novel fusion proteins specific for cd137 and gpc3.
GB201903767D0 (en) 2019-03-19 2019-05-01 Quadrucept Bio Ltd Multimers, tetramers & octamers
KR20210146934A (en) 2019-03-29 2021-12-06 아스트라제네카 에이비이 Lipocalin muteins for the treatment of asthma
CN113939307A (en) 2019-03-29 2022-01-14 皮里斯制药有限公司 Inhalation administration of lipocalin muteins
EP3952996A1 (en) 2019-04-12 2022-02-16 F. Hoffmann-La Roche AG Bispecific antigen binding molecules comprising lipocalin muteins
TW202115124A (en) 2019-06-26 2021-04-16 瑞士商赫孚孟拉羅股份公司 Novel antigen binding molecules binding to cea
WO2020260326A1 (en) 2019-06-27 2020-12-30 F. Hoffmann-La Roche Ag Novel icos antibodies and tumor-targeted antigen binding molecules comprising them
AU2020379176A1 (en) 2019-11-04 2022-06-09 Pieris Pharmaceuticals Gmbh HER2/4-1BB bispecific fusion proteins for the treatment of cancer
WO2021190980A1 (en) 2020-03-22 2021-09-30 Quadrucept Bio Limited Multimers for viral strain evolution
AR121706A1 (en) 2020-04-01 2022-06-29 Hoffmann La Roche OX40 AND FAP-TARGETED BSPECIFIC ANTIGEN-BINDING MOLECULES
WO2021245240A1 (en) 2020-06-05 2021-12-09 Pieris Pharmaceuticals Gmbh Multimeric immunomodulator targeting 4-1bb
JP2023530760A (en) 2020-06-23 2023-07-19 エフ・ホフマン-ラ・ロシュ・アクチェンゲゼルシャフト Agonist CD28 antigen-binding molecules targeting Her2
WO2021260064A1 (en) 2020-06-25 2021-12-30 F. Hoffmann-La Roche Ag Anti-cd3/anti-cd28 bispecific antigen binding molecules
CN117916261A (en) 2020-11-16 2024-04-19 豪夫迈·罗氏有限公司 Combination therapy with FAP-targeted CD40 agonists
KR20230121785A (en) 2020-12-18 2023-08-21 아스트라제네카 에이비이 Lipocalin mutein dry powder formulation for the treatment of asthma
JP2024503826A (en) 2021-01-06 2024-01-29 エフ・ホフマン-ラ・ロシュ・アクチェンゲゼルシャフト Combination therapy using PD1-LAG3 bispecific antibody and CD20 T cell bispecific antibody
US20240101673A1 (en) 2021-02-03 2024-03-28 Mozart Therapeutics, Inc. Binding agents and methods of using the same
WO2022184659A1 (en) 2021-03-01 2022-09-09 Quadrucept Bio Limited Antibody domains & multimers
CA3211591A1 (en) 2021-03-23 2022-09-29 Markus Zettl Her2/4-1bb bispecific fusion proteins for the treatment of cancer
BR112023020859A2 (en) 2021-04-08 2023-12-12 Pieris Pharmaceuticals Gmbh LIPOCALIN MUTEINS SPECIFIC FOR CONNECTIVE TISSUE GROWTH FACTOR (CTGF)
WO2022243341A1 (en) 2021-05-18 2022-11-24 Pieris Pharmaceuticals Gmbh Lipocalin muteins with binding affinity for ox40
WO2022243261A1 (en) 2021-05-19 2022-11-24 F. Hoffmann-La Roche Ag Agonistic cd40 antigen binding molecules targeting cea
AR126009A1 (en) 2021-06-02 2023-08-30 Hoffmann La Roche CD28 ANTIGEN-BINDING AGONIST MOLECULES THAT TARGET EPCAM
WO2023279092A2 (en) 2021-07-02 2023-01-05 Genentech, Inc. Methods and compositions for treating cancer
WO2023076876A1 (en) 2021-10-26 2023-05-04 Mozart Therapeutics, Inc. Modulation of immune responses to viral vectors
WO2023089079A1 (en) 2021-11-19 2023-05-25 Pieris Pharmaceuticals Gmbh Novel fusion protein specific for ox40 and pd-l1
AU2022423749A1 (en) 2021-12-20 2024-06-13 F. Hoffmann-La Roche Ag Agonistic ltbr antibodies and bispecific antibodies comprising them
WO2023186756A1 (en) 2022-03-28 2023-10-05 F. Hoffmann-La Roche Ag Interferon gamma variants and antigen binding molecules comprising these
WO2024030956A2 (en) 2022-08-03 2024-02-08 Mozart Therapeutics, Inc. Cd39-specific binding agents and methods of using the same
WO2024056861A1 (en) 2022-09-15 2024-03-21 Avidicure Ip B.V. Multispecific antigen binding proteins for stimulating nk cells and use thereof
WO2024064713A1 (en) 2022-09-21 2024-03-28 Seagen Inc. Novel fusion protein specific for cd137 and cd228
GB202216503D0 (en) 2022-11-05 2022-12-21 Quadrucept Bio Ltd Non-human vertebrates & cells

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1999016873A1 (en) * 1997-09-26 1999-04-08 Arne Skerra Anticalins
WO2003029463A2 (en) * 2001-09-27 2003-04-10 Pieris Proteolab Ag Muteins of human neutrophil gelatinase-associated lipocalin and related proteins

Family Cites Families (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0677105B1 (en) * 1992-12-31 2006-03-01 Immulogic Pharmaceutical Corporation Allergenic proteins and peptides from dog dander and uses therefor
DE4417598A1 (en) 1994-05-19 1995-12-14 Max Planck Gesellschaft Use of the tetracycline promoter for the stringently regulated production of recombinant proteins in prokaryotic cells
DE19926068C1 (en) 1999-06-08 2001-01-11 Arne Skerra Muteins of the bilin binding protein
US6436703B1 (en) * 2000-03-31 2002-08-20 Hyseq, Inc. Nucleic acids and polypeptides
AU2002255478A1 (en) * 2001-01-10 2002-09-12 Pe Corporation (Ny) Kits, such as nucleic acid arrays, comprising a majority of human exons or transcripts, for detecting expression and other uses thereof
US20030236188A1 (en) * 2001-05-03 2003-12-25 Spytek Kimberly A. Novel human proteins, polynucleotides encoding them and methods of using the same
US7118915B2 (en) 2001-09-27 2006-10-10 Pieris Proteolab Ag Muteins of apolipoprotein D
AU2003275958A1 (en) * 2003-08-25 2005-03-10 Pieris Proteolab Ag Muteins of tear lipocalin

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1999016873A1 (en) * 1997-09-26 1999-04-08 Arne Skerra Anticalins
WO2003029463A2 (en) * 2001-09-27 2003-04-10 Pieris Proteolab Ag Muteins of human neutrophil gelatinase-associated lipocalin and related proteins

Non-Patent Citations (8)

* Cited by examiner, † Cited by third party
Title
GASYMOV OKTAY K ET AL: "Binding studies of tear lipocalin: The role of the conserved tryptophan in maintaining structure, stability and ligand affinity", BIOCHIMICA ET BIOPHYSICA ACTA, vol. 1433, no. 1-2, 17 August 1999 (1999-08-17), pages 307 - 320, XP002269583, ISSN: 0006-3002 *
GASYMOV OKTAY K ET AL: "Relaxation of beta-structure in tear lipocalin and enhancement of retinoid binding", IOVS, vol. 43, no. 10, October 2002 (2002-10-01), pages 3165 - 3173, XP002269582 *
GASYMOV OKTAY K ET AL: "Resolving near-ultraviolet circular dichroism spectra of single trp mutants in tear lipocalin.", ANALYTICAL BIOCHEMISTRY, vol. 318, no. 2, 15 July 2003 (2003-07-15), pages 300 - 308, XP002269551, ISSN: 0003-2697 (ISSN print) *
GASYMOV OKTAY K ET AL: "Site-directed tryptophan fluorescence reveals the solution structure of tear lipocalin: Evidence for features that confer promiscuity in ligand binding", BIOCHEMISTRY, vol. 40, no. 49, 11 December 2001 (2001-12-11), pages 14754 - 14762, XP002269552, ISSN: 0006-2960 *
REDL BERNHARD: "Human tear lipocalin", BIOCHIMICA ET BIOPHYSICA ACTA, vol. 1482, no. 1-2, 18 October 2000 (2000-10-18), pages 241 - 248, XP002269555, ISSN: 0006-3002 *
SCHLEHUBER S ET AL: "DUOCALINS: ENGINEERED LIGAND-BINDING PROTEINS WITH DUAL SPECIFICITY DERIVED FROM THE LIPOCALIN FOLD", BIOLOGICAL CHEMISTRY, XX, XX, vol. 382, no. 9, September 2000 (2000-09-01), pages 1335 - 1342, XP001078830, ISSN: 1431-6730 *
SKERRA A: "'Anticalins': a new class of engineered ligand-binding proteins with antibody-like properties.", JOURNAL OF BIOTECHNOLOGY. NETHERLANDS JUN 2001, vol. 74, no. 4, June 2001 (2001-06-01), pages 257 - 275, XP002269554, ISSN: 0168-1656 *
SKERRA A: "ENGINEERED PROTEIN SCAFFOLDS FOR MOLECULAR RECOGNITION", JOURNAL OF MOLECULAR RECOGNITION, HEYDEN & SON LTD., LONDON, GB, vol. 13, no. 4, July 2000 (2000-07-01), pages 167 - 187, XP009019725, ISSN: 0952-3499 *

Cited By (74)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2899277A1 (en) 2004-11-26 2015-07-29 Pieris AG Compound with affinity for the cytotoxic T lymphocyte-associated antigen (CTLA-4)
US7611847B2 (en) 2005-06-01 2009-11-03 Agency For Science, Technology And Research Method for identifying an intestinal phenotype
US8216855B2 (en) 2006-02-13 2012-07-10 Agency For Science, Technology And Research Method of processing a biological and/or chemical sample
US8261598B2 (en) 2006-03-09 2012-09-11 Agency For Science, Technology And Research Apparatus for performing a reaction in a droplet and method of using the same
WO2008015239A2 (en) 2006-08-01 2008-02-07 Pieris Ag Muteins of tear lipocalin and methods for obtaining the same
US9581527B2 (en) 2006-11-24 2017-02-28 Agency For Science, Technology And Research Apparatus for processing a sample in a liquid droplet and method of using the same
WO2008063136A1 (en) 2006-11-24 2008-05-29 Agency For Science, Technology And Research Apparatus for processing a sample in a liquid droplet and method of using the same
US8691147B2 (en) 2006-11-24 2014-04-08 Agency For Science, Technology And Research Apparatus for processing a sample in a liquid droplet and method of using the same
US9874501B2 (en) 2006-11-24 2018-01-23 Curiox Biosystems Pte Ltd. Use of chemically patterned substrate for liquid handling, chemical and biological reactions
US8663915B2 (en) 2007-04-13 2014-03-04 Agency For Science, Technology And Research Methods of controlling tumorigenesis and diagnosing the risk thereof
US8969313B2 (en) 2007-11-02 2015-03-03 Agency For Science, Technology And Research Methods and compounds for preventing and treating a tumour
US10725020B2 (en) 2007-11-14 2020-07-28 Curiox Biosystems Pte Ltd. High throughput miniaturized assay system and methods
US9102920B2 (en) 2008-05-06 2015-08-11 Agency For Science, Technology And Research Method of effecting de-differentiation of a cell
EP3381933A1 (en) 2008-06-24 2018-10-03 Technische Universität München Muteins of hngal and related proteins with affinity for a given target
US8716420B2 (en) 2008-10-13 2014-05-06 Agency For Science, Technology And Research Amphiphilic polymers and nanocrystals coated therewith
EP2189539A1 (en) 2008-11-21 2010-05-26 Chimera Biotec GmbH Conjugate complexes for analyte detection
US8927210B2 (en) 2008-11-21 2015-01-06 Chimera Biotec Gmbh Conjugate complexes for analyte detection
US8784752B2 (en) 2009-04-17 2014-07-22 Curiox Biosystems Pte Ltd Use of chemically patterned substrate for liquid handling, chemical and biological reactions
US8871686B2 (en) 2009-07-07 2014-10-28 Agency For Science, Technology And Research Methods of identifying a pair of binding partners
EP2550958A1 (en) 2009-08-05 2013-01-30 Pieris AG Controlled release formulations of lipocalin muteins
WO2011015634A2 (en) 2009-08-05 2011-02-10 Pieris Ag Controlled release formulations of lipocalin muteins
US11827681B2 (en) 2009-12-07 2023-11-28 Pieris Pharmaceuticals Gmbh Muteins of human lipocalin 2 (Lcn2, hNGAL) with affinity for a given target
US10618941B2 (en) 2009-12-07 2020-04-14 Pieris Pharmaceuticals Gmbh Muteins of human lipocalin 2 (Lcn2,hNGAL) with affinity for a given target
EP3660510A2 (en) 2009-12-07 2020-06-03 Pieris Pharmaceuticals GmbH Muteins of human lipocalin 2 (lcn2, hngal) with affinity for a given target
US9549968B2 (en) 2009-12-07 2017-01-24 Pieris Pharmaceuticals Gmbh Muteins of human lipocalin 2 (LcnC,hNGAL) with affinity for a given target
EP2990798A1 (en) 2009-12-07 2016-03-02 Pieris AG Muteins of human lipocalin 2 (lcn2, hngal) with affinity for a given target
WO2011069992A2 (en) 2009-12-07 2011-06-16 Pieris Ag Muteins of human lipocalin 2 (lcn2, hngal) with affinity for a given target
WO2011078794A1 (en) 2009-12-22 2011-06-30 Agency For Science, Technology And Research Sers-based analyte detection
US9689801B2 (en) 2009-12-22 2017-06-27 Agency For Science, Technology And Research SERS-based analyte detection
US8986951B2 (en) 2010-06-08 2015-03-24 Pieris Ag Tear lipocalin in muteins binding IL-4 R alpha
US10232014B2 (en) 2010-06-08 2019-03-19 Pieris Pharmaceuticals Gmbh Tear lipocalin muteins binding IL-4 R alpha
US10898545B2 (en) 2010-06-08 2021-01-26 Astrazeneca Ab Tear lipocalin muteins binding IL-4 r alpha
WO2011154420A2 (en) 2010-06-08 2011-12-15 Pieris Ag Tear lipocalin muteins binding il-4 r alpha
US9687524B2 (en) 2010-06-08 2017-06-27 Pieris Pharmaceuticals Gmbh Tear lipocalin muteins binding IL-4 R alpha
US10632468B2 (en) 2010-07-23 2020-04-28 Curiox Biosystems Pte Ltd. Apparatus and method for multiple reactions in small volumes
US9878328B2 (en) 2010-07-23 2018-01-30 Curiox Biosystems Pte Ltd. Apparatus and method for multiple reactions in small volumes
WO2012022742A1 (en) 2010-08-16 2012-02-23 Pieris Ag Binding proteins for hepcidin
US9051382B2 (en) 2010-08-16 2015-06-09 Pieris Ag Human neutrophil gelatinase-associated lipocalin (hNGAL) muteins that bind hepcidin and nucleic acid encoding such
EP3299386A1 (en) 2010-08-16 2018-03-28 Pieris Pharmaceuticals GmbH Binding proteins for hepcidin
WO2012065978A1 (en) 2010-11-15 2012-05-24 Pieris Ag Muteins of human lipocalin 2 with affinity for glypican-3 (gpc3)
US9260492B2 (en) 2010-11-15 2016-02-16 Pieris Ag Muteins of human lipocalin 2 with affinity for glypican-3 (GPC-3)
WO2012072806A1 (en) 2010-12-02 2012-06-07 Pieris Ag Muteins of human lipocalin 2 with affinity for ctla-4
US9221885B2 (en) 2010-12-02 2015-12-29 Pieris Ag Muteins of human lipocalin 2 with affinity for CTLA-4
US10857202B2 (en) 2011-12-13 2020-12-08 Pieris Pharmaceuticals Gmbh Methods for preventing or treating certain disorders by inhibiting binding of IL-4 and/or IL-13 to their respective receptors
US11382951B2 (en) 2011-12-13 2022-07-12 Pieris Pharmaceuticals Gmbh Methods for preventing or treating certain disorders by inhibiting binding of IL-4 and/or IL-13 to their respective receptors
US10398754B2 (en) 2011-12-13 2019-09-03 Pieris Pharmaceuticals Gmbh Methods for preventing or treating certain disorders by inhibiting binding of IL-4 and/or IL-13 to their respective receptors
US9950323B2 (en) 2012-02-05 2018-04-24 Curiox Biosystems Pte Ltd. Array plates and methods for making and using same
US10792661B2 (en) 2012-02-05 2020-10-06 Curiox Biosystems Pte Ltd. Array plates and methods for making and using same
WO2013174783A1 (en) 2012-05-23 2013-11-28 Pieris Ag Lipocalin muteins with binding-affinity for glypican-3 (gpc-3) and use of lipocalin muteins for target-specific delivery to cells expressing gpc-3
WO2013189521A1 (en) 2012-06-19 2013-12-27 Waclawczyk Simon Method of generating cells of hepatocyte phenotype
US10526384B2 (en) 2012-11-19 2020-01-07 Pieris Pharmaceuticals Gmbh Interleukin-17A-specific and interleukin-23-specific binding polypeptides and uses thereof
JP2016519051A (en) * 2013-03-14 2016-06-30 第一三共株式会社 Novel PCSK9 binding protein
US9150629B2 (en) 2013-03-14 2015-10-06 Daiichi Sankyo Co., Ltd. Human tear lipocalins which bind PCSK9 and methods of use thereof
WO2014140210A1 (en) 2013-03-14 2014-09-18 Pieris Ag Novel binding proteins for pcsk9
US9598476B2 (en) 2013-03-14 2017-03-21 Daiichi Sankyo Co., Ltd. Nucleic acid molecules encoding muteins of human tear lipocalin which bind PCSK9
US9557318B2 (en) 2013-07-09 2017-01-31 Curiox Biosystems Pte Ltd. Array plates for washing samples
US10927154B2 (en) 2014-01-13 2021-02-23 Pieris Pharmaceuticals Gmbh Multi-specific polypeptide useful for localized tumor immunomodulation
WO2015140314A1 (en) 2014-03-21 2015-09-24 Philipps-Universität Marburg Means and methods for itaconic acid production
US10774119B2 (en) 2014-05-22 2020-09-15 Pieris Pharmaceuticals Gmbh Specific-binding polypeptides and uses thereof
US11345728B2 (en) 2014-05-22 2022-05-31 Pieris Pharmaceuticals Gmbh Specific-binding polypeptides and uses thereof
US10526382B2 (en) 2015-01-28 2020-01-07 Pieris Pharmaceuticals Gmbh Human neutrophil gelatinase-associated lipocalin (hNGAL) muteins capable of binding angiopoietin-2 (Ang-2) and methods of use thereof
US11034738B2 (en) 2015-01-28 2021-06-15 Pieris Pharmaceuticals Gmbh Nucleic acid molecules encoding human neutrophil gelatinase-associated lipocalin (hNGAL) which bind angiopoietin-2 (Ang-2)
WO2016142314A1 (en) 2015-03-06 2016-09-15 Deutsches Krebsforschungszentrum FUSION PROTEINS COMPRISING A BINDING PROTEIN AND AN INTERLEUKIN-15 POLYPEPTIDE HAVING A REDUCED AFFINITY FOR IL15Ra AND THERAPEUTIC USES THEREOF
US11261221B2 (en) 2015-05-04 2022-03-01 Pieris Pharmaceuticals Gmbh Proteins specific for CD137
US10865250B2 (en) 2015-05-04 2020-12-15 Pieris Pharmaceuticals Gmbh Anti-cancer fusion polypeptide
US10787491B2 (en) 2015-05-18 2020-09-29 Pieris Pharmaceuticals Gmbh Nucleic acid molecules encoding muteins of human lipocalin 2 with affinity for glypican-3 (GPC3)
WO2016184875A1 (en) 2015-05-18 2016-11-24 Pieris Pharmaceuticals Gmbh Muteins of human lipocalin 2 with affinity for glypican-3 (gpc3)
US10913778B2 (en) 2015-05-18 2021-02-09 Pieris Pharmaceuticals Gmbh Anti-cancer fusion polypeptides, encoding nucleic acids and methods of using polypeptides
US10545139B2 (en) 2015-06-16 2020-01-28 Curiox Biosystems Pte Ltd. Methods and devices for performing biological assays using magnetic components
US10703810B2 (en) 2015-11-30 2020-07-07 Pieris Australia Pty Ltd. Fusion polypeptides which bind vascular endothelial growth factor a (VEGF-A) and angiopoietin-2 (Ang-2)
US11034737B2 (en) 2015-12-10 2021-06-15 Pieris Pharmaceuticals Gmbh Proteins specific for calcitonin gene-related peptide
US10400016B2 (en) 2015-12-10 2019-09-03 Pieris Pharmaceuticals Gmbh Proteins specific for calcitonin gene-related peptide
US11692162B2 (en) 2017-04-05 2023-07-04 Curiox Biosystems Pte Ltd. Methods, devices, and apparatus for washing samples on array plates
WO2019243564A1 (en) 2018-06-21 2019-12-26 Technische Universitaet Muenchen Hepatitis b and/or hepatitis d-permissive cells and animals

Also Published As

Publication number Publication date
WO2005019256A2 (en) 2005-03-03
CA2536491C (en) 2013-12-24
US20070224633A1 (en) 2007-09-27
US20090325875A1 (en) 2009-12-31
SG131109A1 (en) 2007-04-26
DK1660528T3 (en) 2010-01-04
US7893208B2 (en) 2011-02-22
AU2004266846A1 (en) 2005-03-03
BRPI0413728A (en) 2006-10-24
HK1084404A1 (en) 2006-07-28
JP2007531503A (en) 2007-11-08
CN1871256B (en) 2010-06-23
AU2004266846B2 (en) 2011-01-06
EP1660528A2 (en) 2006-05-31
DE602004023725D1 (en) 2009-12-03
AU2003275958A1 (en) 2005-03-10
ATE446313T1 (en) 2009-11-15
CN1871256A (en) 2006-11-29
JP4856543B2 (en) 2012-01-18
US7585940B2 (en) 2009-09-08
WO2005019256A3 (en) 2005-07-21
BRPI0413728B8 (en) 2021-05-25
WO2005019255A8 (en) 2005-05-26
CA2536491A1 (en) 2005-03-03
BRPI0413728B1 (en) 2017-05-23
EP1660528B1 (en) 2009-10-21

Similar Documents

Publication Publication Date Title
WO2005019255A1 (en) Muteins of tear lipocalin
US7691970B2 (en) Muteins of a bilin-binding protein with affinity for a given target
AU2002337111B2 (en) Muteins of human neutrophil gelatinase-associated lipocalin and related proteins
US7118915B2 (en) Muteins of apolipoprotein D
CA2583009C (en) Ubiquitin or gamma-crystalline conjugates for use in therapy, diagnosis and chromatography
US7250297B1 (en) Anticalins
US5723584A (en) Biotinylation of proteins
AU2002337111A1 (en) Muteins of human neutrophil gelatinase-associated lipocalin and related proteins
US20160280747A1 (en) Anticalins
US10738092B2 (en) Muteins of a1m lipocalin and method of production therefor

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SY TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IT LU MC NL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
CFP Corrected version of a pamphlet front page
CR1 Correction of entry in section i

Free format text: IN PCT GAZETTE 09/2005 UNDER (81) DESIGNATIONS SHOULD READ "US" AND UNDER (84) DESIGNATIONS SHOULD READ "US"

NENP Non-entry into the national phase

Ref country code: JP

WWW Wipo information: withdrawn in national office

Country of ref document: JP

122 Ep: pct application non-entry in european phase