WO1995019374A1 - Bacterial receptor structures - Google Patents

Bacterial receptor structures Download PDF

Info

Publication number
WO1995019374A1
WO1995019374A1 PCT/SE1995/000034 SE9500034W WO9519374A1 WO 1995019374 A1 WO1995019374 A1 WO 1995019374A1 SE 9500034 W SE9500034 W SE 9500034W WO 9519374 A1 WO9519374 A1 WO 9519374A1
Authority
WO
WIPO (PCT)
Prior art keywords
proteins
protein
receptor
bacterial
receptors
Prior art date
Application number
PCT/SE1995/000034
Other languages
French (fr)
Other versions
WO1995019374A9 (en
Inventor
Björn Nilsson
Per-Åke Nygren
Mathias Uhlén
Original Assignee
Pharmacia Ab
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 Pharmacia Ab filed Critical Pharmacia Ab
Priority to EP95907175A priority Critical patent/EP0739353B1/en
Priority to JP51898495A priority patent/JP4089920B2/en
Priority to DE69533644T priority patent/DE69533644T2/en
Priority to AU15487/95A priority patent/AU696186B2/en
Priority to CA002181042A priority patent/CA2181042C/en
Priority to DK95907175T priority patent/DK0739353T3/en
Priority to AT95907175T priority patent/ATE279439T1/en
Priority to US08/669,360 priority patent/US5831012A/en
Publication of WO1995019374A1 publication Critical patent/WO1995019374A1/en
Publication of WO1995019374A9 publication Critical patent/WO1995019374A9/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/10Processes for the isolation, preparation or purification of DNA or RNA
    • C12N15/1034Isolating an individual clone by screening libraries
    • C12N15/1037Screening libraries presented on the surface of microorganisms, e.g. phage display, E. coli display
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/195Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from bacteria
    • C07K14/305Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from bacteria from Micrococcaceae (F)
    • C07K14/31Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from bacteria from Micrococcaceae (F) from Staphylococcus (G)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/195Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from bacteria
    • C07K14/315Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from bacteria from Streptococcus (G), e.g. Enterococci
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10STECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10S435/00Chemistry: molecular biology and microbiology
    • Y10S435/8215Microorganisms
    • Y10S435/822Microorganisms using bacteria or actinomycetales
    • Y10S435/882Staphylococcus
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10STECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10S435/00Chemistry: molecular biology and microbiology
    • Y10S435/8215Microorganisms
    • Y10S435/822Microorganisms using bacteria or actinomycetales
    • Y10S435/882Staphylococcus
    • Y10S435/883Staphylococcus aureus
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10STECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10S530/00Chemistry: natural resins or derivatives; peptides or proteins; lignins or reaction products thereof
    • Y10S530/82Proteins from microorganisms
    • Y10S530/825Bacteria

Definitions

  • Fc receptors Several bacteria known to invade mammals have evolved surface proteins capable of binding to a variety of sub ⁇ stances including host specific carbohydrates and prote ⁇ ins. Several such receptors from Gram-positive bacterial pathogens have been isolated and characterized in detail as will be shown below. Most well-characterized are the Fc receptors, named after the capability of binding to the constant Fc part of IgG. Based on binding experiments to IgG from various mammalian sources, and subclasses there- of, Fc receptors have been divided into six types I-VI.
  • SPA protein A
  • the structure for a complex between human Fc [IgGl] and a single domain [B] of SPA has been determined by X- ray crystallography at a resolution of 2.8 A [Deisenhofer, J. et al 1981 Biochemistry 20:2361-2370]. Based on this structure and additional information from NMR experiments, the B domain can be viewed as a compact structure consis- ting of three anti-parallel -helices connected with loops. In the Fc binding, which is of both electrostatic and hydrophobic nature, only side chains of residues from helices 1 and 2 are involved, whilst the third helix is not participating in the binding. Based on this domain B, a synthetic IgG-binding domain [Z] [Nilsson, B.
  • Streptococcal strains of serological groups C and G display a binding repertoire for mammalian IgGs, including human IgG3, which is even broader than for the type I re ⁇ ceptor.
  • the name protein G was suggested for this type III receptor from group G streptococci.
  • Olsson and co- workers reported on the cloning and sequencing of the gene from the serological group G streptococci [G148] [Guss, B. et al, 1987 EMBO J. 5:1567-1575; Olsson, A. et al, 1987 Eur.J.Biochem. 168:319-324].
  • SPG In analogy with SPA is SPG a repetitively arranged molecule, comprising an IgG-binding region of three homologous domains [C1,C2,C3], spaced by smaller D-regions (Fig. 2A). Compared to SPA, SPG displays a different binding spectra for imrnunoglobulins from dif ⁇ ferent species and subclasses thereof.
  • the IgG binding domains of protein G are now widely used as an immuno- logical tool, i.e. in the affinity purification of mono ⁇ clonal antibodies. Production of subfragments constructed by DNA-technology, have shown that an individual C-region is sufficient for full IgG-binding.
  • SPG serum albumin binding strength
  • the binding strength is species depen- dent, and among the tested samples, SPG binds strongest to serum albumin from rat, man and mouse.
  • Production and bin ⁇ ding studies of subfragments of SPG shows that the two binding activities are structurally separated and that the serum albumin binding function is located at the repeti- tive A-B region [Nygren et al 1990 Eur.J.Biochem. 193:143- 148] . This region has been used for several biotechnolo- gical purposes. Recombinant proteins have been produced as fusions to the region which enables the purification by affinity chroma ography, where human serum albumin most frequently has been used as immobilized ligand.
  • Proteins found to be proteolytically sensitive have been produced as "dual affinity fusions" in which they are flanked by two different affinity tails derived from SPA and SPG, respectively. Purification schemes employing both the N- and C-terminal are thus possible which ensures the recove ⁇ ry of an intact target protein [Hammarberg et al 1989 Proc.Natl.Acad.Sciences USA 86:4367-4371], The strong and specific binding to serum albumin has also been used for the In vivo stabilization of therapeutic proteins.
  • the receptor is carried in the circulation (macaque apes) with a half-life which is close to the half-life for serum albumin itself.
  • mice with the for HIV/AIDS therapy interesting, but rapidly cleared T-cell receptor CD4 showed that it was substantially sta ⁇ bilized when fused to the serum albumin binding region, when compared with an unfused control protein [Nygren et al 1991 Vaccines 91 Cold Spring Harbor Press 363-368] .
  • the slow clearance can probably be explained by the complex formation with serum albumin which circumvents elimination by the liver and excretion in the kidney.
  • SPG is regarded to be trivalent with regard to binding to serum albumin. Similar to the monovalent IgG-binding domains Z and Cl B2A3 is relatively small and shows high solubility and stability and is therefore a suitable candidate for modification.
  • the present invention has for its main purpose to provide new bacterial receptor structures by modifying natural bacterial receptors in regard to their original interaction functions to result in new structures having modified interaction functions.
  • Another object of the invention is to provide arti ⁇ ficial bacterial receptor structures which are stable and more resistant to various conditions, such as increased temperatures.
  • Yet another object of the invention is to provide artificial bacterial receptor structures, the interaction functions of which have been modified to direct same to other desired interaction partners.
  • novel proteins obtainable by mutagenesis of surface- exposed amino acids of domains of natural bacterial re- ceptors said proteins being obtained without substantial loss of basic structure and stability of said natural bacterial receptors.
  • Said proteins have preferably been selected from a protein library embodying a repertoire of said novel proteins.
  • at least one amino acid residue involved in the interaction fuction of the or._jinal bacterial receptor has been made subject to substitution by another amino acid residue so as to result in substantial loss of the original interaction capacity with a modified interaction capacity being created, said substitution being made with ⁇ out substantial loss of basic structure and stability of the original bacterial receptor.
  • said bacterial structures origi ⁇ nate from Gram-positive bacteria there may be mentioned Staphylococcu ⁇ aureus .
  • Streptococcus pyogenes [group A] Streptococcus group C,G,L, bovine group G streptococci, Streptococcus zooepidemicus [group C] , Streptococcus zooepidemicu ⁇ S212, Streptococcus pyogenes [group A] , streptococci groups A,C,G, Peptostreptococcus magnus, Streptococcus agalactiae [group B] .
  • thermophilic bacteria evolved to persist in environments of elevated temperatures.
  • Re ⁇ ceptors from species like e.g. Bacillus ⁇ tearother ⁇ o- philu ⁇ , Ther u ⁇ aquaticu ⁇ , Theirmococcu ⁇ litoralis and Pyrococcus have the potential of being naturally excep ⁇ tionally stable, thus suitable for providing structural frameworks for protein engineering according to the in ⁇ vention.
  • bac ⁇ terial receptors originating from Fc[IgG]receptor type I, type II, type III, type IV, type V and type VI, fibronec- tin receptor, M protein, plasmin receptor, collagen re ⁇ ceptor, fibrinogen receptor or protein L [K light chains], protein H [human IgG], protein B [human IgA,Al], protein Arp [human IgA] .
  • Particularly preferred bacterial receptors originate from the Fc[IgG]receptor type I of staphylococcal protein A or the serum albumin receptor of streptococcal protein G.
  • the substi ⁇ tution involving amino acid residues taking part in the interaction function of the original bacterial receptor does not involve more than about 50% of the amino acid residues of the original bacterial receptor. It is parti- cularly preferred that not more than about 25% of the amino acid residues of the original bacterial receptor are made subject to substitution.
  • substitution thereof involves not more than substantially all of the amino acid residues taking part in the interaction function of the original bacterial receptor.
  • the bacterial receptor according to the pre ⁇ sent invention is comprised of not more than about 100 amino acid residues. It is known from scientific reports that proteins of a relatively small size are fairly resis ⁇ tant to increased temperatures and also to low pH and cer- tain chemicals. For details concerning temperature resis ⁇ tance c.f. the article by Alexander et al. in Biochemistry 1992, 31, pp 3597-3603.
  • interaction part ⁇ ners are IGF-I, IGF-II, hGH, Factor VIII, insulin and apolipoprotein and their respective receptors as interac ⁇ tion partners. Furthermore, by selecting new receptor variants with specificity for different folding forms of proteins, affinity resins or analytical tools to facili ⁇ tate the isolation of correctly folded molecules can be produced. Further examples are viral coat proteins, bac ⁇ terial antigens, biotin and cell markers, such as CD 34 and CD 4.
  • the present invention is applicable to a variety of natural bacterial receptors the following illustration of the invention more in detail will be directed to the use of the IgG-binding domains Z, Cl and B2A3.
  • the concept of the present invention residing in the use of artificial bacterial receptors based on the natural structures of naturally occurring bacterial receptors is associated with several advantages.
  • the invention makes it possible to use robust and stable, highly soluble and secretion competent receptors. This is in contrast to previous techniques based on the use of polyclonals and monoclonals, such as for diagnostic purposes, which are not very stable in connection with storage, varying condi ⁇ tions, such as varying temperatures etc.
  • the invention makes it possible to modify natural bacterial receptors to obtain desired interaction capacities for specific purposes.
  • phage display technique can be used for selection of both small peptides as well as more complicated proteins such as antibodies, receptors and hormones.
  • intra- cellular systems have been developed in which the library of proteins are fused to a repressor protein with affinity for a specific plasmid-borne operator region resulting in a coupling between the specific protein variant and the plasmid that encoded it.
  • An alternative to the phages as bearer of protein libraries would be to use bacterial cells.
  • bacterial recep ⁇ tors as structural frameworks are several. They have evol ⁇ ved to express a binding function without disturbing the overall structure. They are naturally highly soluble, robust to unphysiological conditions, such as pH and heat, folding efficient and are in addition secretion competent. The invention finds use in several different areas.
  • the introductory part of the above-identified patent specification WO92/20791 gives an excellent survey on an ⁇ tibodies and their structure. Reference is particularly made to page 1 thereof.
  • the bacterial receptors SPA and SPG have been widely used in antibody technology for detection and purification of antibodies from e.g. hybridom supernatants and ascites fluids. However, not all antibodies are recognized by these receptors, depending on species and subclass. For the smaller subfragments of antibodies (Fig. 4), SPA and SPG show a limited binding, and efficient tools for gene- ral purification schemes are lacking. However, from a repertoire of mutant receptors including SPA and SPG, forms displaying a broader affinity for antibodies and subfragments thereof can potentially be selected.
  • the complex structural organization of antibodies has a number of consequences for their use in different appli ⁇ cations as well for the production of recombinant deriva ⁇ tives.
  • the arrangement of sub- units connected by disulphide bonds can lead to a leakage of released heavy and light chains from columns.
  • the re ⁇ quirement of successful docking of the two subunits con ⁇ tributing to the antigen binding site makes the production in bacteria of small subfragments with a low association difficult.
  • the folding of the antibody is dependent on the formation of intra- and inter chain disulphidebonds, which are not able to form in the intracellular environment of bacterial cells.
  • High-level intracellular expression sys ⁇ tems for recombinant antibodies leads to the formation of inclusion bodies, which have to be renatured to obtain biological activity.
  • the CDR regions forming the antigen bidning part of an antibody forms a total surface available for the anti ⁇ gen of approximately 800 A 2 , with typical 10-20 residues from the antibody involved in the binding.
  • the binding surface of about 600 A 2 is of the same order of magnitude as between an antibody and its antigen.
  • Immunoglobulins are substantially tetramers built up from so called ⁇ -sheet structures which stabilize the orientation of the antigen- binding loops which in turn consist of continuous peptide sequences.
  • the purification of the product is frequently a major problem.
  • the hybrid product can effectively and selec ⁇ tively be recovered from the cell lysate or in certain cases from the culture medium by passage through a column containing an immobilized ligand.
  • gene fusion systems have been described which are based on the inter- action of a certain protein with a ligand. For industrial applications it is often desirable to clean effectively the columns between the runs to satisfy purity require ⁇ ments by authorities.
  • the relatively harsh conditions NaOH, acids, heat
  • organic or physical matrises for example in ion exchange chromatography and gel filtration
  • the use of new ligands based on stable structures originating from bacterial receptors are of great importance.
  • the Z domain from SPA is an excellent example since said domain can be subjected to such difficult conditions as a pH of 1 or heating to 80°C without denaturating non-reversibly (see Example 2 below).
  • Z variants inte ⁇ resting protein products can be selected for use immobi ⁇ lized on a solid phase for affinity chromatography.
  • the high solubility of the said domain enables the use of in ⁇ creased solubility of proteins in either refolding from inclusion bodies or in so called reshuffling of disulphide bridges.
  • new forms can be selected having improved properties to facilitate and even make refolding of recombinant proteins possible (cis-acting chaperones) .
  • Recently a new unit operation for the purification of recombinant proteins based on ion exchange chromatography in so called expanded bed has been described [Hansson, M. et al 1994 Bio/Technol. in press].
  • the present invention provides means for producing and selecting proteins with novel functions. According to the invention this is achieved by extensive mutation of defined residues of stable domains of bacterial receptors. Due to the novel functions of the artificial bacterial re ⁇ ceptors, these can be used as specific binders for thera ⁇ Illustrated, diagnostic, biotechnology or in research. 17 Figure 9. Phagemid vector pKNl .
  • the library PCR products encoding the variegated helices 1 and 2 was subclo- ned into the phagemid vector, pKNl, containing the gene for residues 44-58 of the wild type Z domain (essentially helix 3), followed by the gene for a 46 residues serum albumin binding region (ABP) derived from streptococcal protein G linked in frame with a truncated version of the M13 phage coat protein 3 gene.
  • the phagemid contains the origin of replication derived from plasmid pBR322 as well as the intergenic region ( f1 ori ) required for packing into the phage particles.
  • FIG. 10 SDS-PAGE. HSA-affinity purified proteins from the periplasm of E ⁇ cherichia coli cells produ ⁇ cing the wild type Z domain and two different acid Z-variants as ABP fusion proteins encoded from their respective phagemid vectors were analyzed by SDS/PAGE.
  • M molecular weight mar ⁇ ker; lane 1, wild type Z domain; lane 2, clone 10; lane 3, clone 12.
  • FIG. 11 CD-data. Overlay plot of CD spectra obtained for the wild type Z domain and two variants of the Z-protein library. The signals of the pro ⁇ teins were obtained after subtraction of the CD signal contribution of the ABP tail, present during the analysis.
  • FIG. 12 Ion exchange chromatography.
  • the two acid Z- variant proteins no. 10 and no. 12 together with the wild type Z-domain (produced as ABP fusion proteins) were each subjected to analy ⁇ sis at pH 5.5, employing an anion exchange chromatography column. Elution of the proteins 18 from the column was obtained by a NaCl gradi ⁇ ent.
  • FIG. 13 Z-domain structure.
  • the structure of helices one and two are from the co-crystal structure be ⁇ tween domain B of SPA and Fc (Deisenhofer, (1981) Biochemistry, 20, 2361-2370).
  • the third helix was built based on the secondary structu ⁇ re assignments from NMR spectroscopy (Gouda et al., (1992) Biochemistry, 31, 9665-9672).
  • Non- hydrogen atoms of side-chains of residues that were mutated in the construction of the combi ⁇ natorial library are displayed as ball-and- stick models.
  • the display was generated by the program MOLSCRIPT (Kraulis (1991) J.Appl.
  • Figure 14 Amino acid sequences. Result from DNA-sequen- cing of 31 randomly chosen Z-variants from the library. The residues subjected to the mutage ⁇ nesis are boxed. Horisontal lines indicate nu- cleotide identity with the wild type Z sequence listed at the top. Indicated are the clones that were expressed and characterized as fusion proteins to the ABP-tail.
  • Figure 15 Aminoacid distribution. Result from the statis ⁇ tical analysis of the deduced amino acids at the mutated positions. In total, 13 residues from 31 clones (403 codons) were included in the calculation. The ratios between observed and expected frequencies are shown for all 20 Figure 9 Phagemid vector pKNl .
  • the library PCR products encoding the variegated helices 1 and 2 was subclo- ned into the phagemid vector, pKNl, containing the gene for residues 44-58 of the wild type Z domain (essentially helix 3), followed by the gene for a 46 residues serum albumin binding region (ABP) derived from streptococcal protein G linked in frame with a truncated version of the M13 phage coat protein 3 gene.
  • the phagemid contains the origin of replication derived from plasmid pBR322 as well as the intergenic region ( f1 ori ) required for packing into the phage particles.
  • FIG. 10 SDS-PAGE. HSA-affinity purified proteins from the periplasm of E ⁇ cherichia coli cells produ ⁇ cing the wild type Z domain and two different acid Z-variants as ABP fusion proteins encoded from their respective phagemid vectors were analyzed by SDS/PAGE.
  • M molecular weight mar ⁇ ker; lane 1, wild type Z domain; lane 2, clone 10; lane 3, clone 12.
  • FIG. 11 CD-data. Overlay plot of CD spectra obtained for the wild type Z domain and two variants of the Z-protein library. The signals of the pro ⁇ teins were obtained after subtraction of the CD signal contribution of the ABP tail, present during the analysis.
  • FIG 12 Ion exchange chromatography.
  • the two acid Z- variant proteins no. 10 and no. 12 together with the wild type Z-domain (produced as ABP fusion proteins) were each subjected to analy ⁇ sis at pH 5.5, employing an anion exchange chromatography column. Elution of the proteins from the column was obtained by a NaCl gradi ⁇ ent.
  • FIG. 13 Z-domain structure.
  • the structure of helices one and two are from the co-crystal structure be ⁇ tween domain B of SPA and Fc (Deisenhofer, (1981) Biochemistry, 20, 2361-2370).
  • the third helix was built based on the secondary structu ⁇ re assignments from NMR spectroscopy (Gouda et al., (1992) Biochemistry, 31, 9665-9672).
  • Non- hydrogen atoms of side-chains of residues that were mutated in the construction of the combi ⁇ natorial library are displayed as ball-and- stick models.
  • the display was generated by the program MOLSCRIPT (Kraulis (1991) J.Appl.
  • Figure 14 Amino acid sequences. Result from DNA-sequen- cing of 31 randomly chosen Z-variants from the library. The residues subjected to the mutage ⁇ nesis are boxed. Horisontal lines indicate nu- cleotide identity with the wild type Z sequence listed at the top. Indicated are the clones that were expressed and characterized as fusion proteins to the ABP-tail.
  • Figure 15 Aminoacid distribution. Result from the statis ⁇ tical analysis of the deduced amino acids at the mutated positions. In total, 13 residues from 31 clones (403 codons) were included in the calculation. The ratios between observed and expected frequencies are shown for all 20 amino acids as well for the only termination signal (TAG) included in the NNG/T degeneracy profile.
  • TAG termination signal
  • FIG. 16 SDS-PAGE analysis. HSA-affinity purified pro ⁇ teins from the periplasm of E.coli cells produ ⁇ cing the wild type Z domain and four different Z-variants as ABP fusion proteins encoded from their respective phagemid vectors were analyzed by SDS/PAGE. Lanes 1-5: Reduced conditions.
  • Lanes 6 and 7 Non-reduced conditions. Lane 1, wild type Z domain; lane 2, clone 16; lane 3, clone 21; lane 4, clone 22; lane 5, clone 24; M, molecular weight marker; lane 6, clone 16 and lane 7, clone 22.
  • Figure 17 CD-data. Overlay plot of CD spectra obtained for the wild type Z domain and four variants of the ⁇ -helical protein surface library. The sig- nals of the variants were obtained after sub ⁇ traction of the CD signal contribution of the ABP tail, present during the analysis.
  • FIG. 18 Biosensor assay. An overlay plot of sensorgrams obtained from the BIA-coreTM analysis of the wild type Z domain and four different variants (no. 16,21,22,24; Figure 4) fused to the ABP tail. The IgG-binding activities of the diffe ⁇ rent proteins were analyzed using a sensor chip coated with approx. 5000 RU human polyclonal
  • oligonucleotides (Fig. 6) were purchased from Scandinavian Gene Synthesis (Sweden), and phosphorylated where indicated according to [Maniatis et al (1988) Mole ⁇ cular cloning. A laboratory manual. Cold Spring Harbor Laboratory Press].
  • ZLIB-1 was biotinylated in the 5 ' -end enabling immobilization on paramagnetic beads M-280 Strep- tavidin purchased from Dynal A/S (Norway). Washing/binding buffer was 1 M NaCl, 10 mM Tris-HCl, pH 7.5, 1 mM EDTA (ethylenediamine tetraacetic acid).
  • the annealing/ligation buffer was 30 mM Tris-HCl, pH 7.5, 10 mM MgCl 2 , 0.2 mM ATP, 1 mM 1.4 dithiothreitol (DTT).
  • DNA ligase were from Boehringer Mannheim, Germany.
  • 10 x PCR buffer contained 20 rnM MgCl 2 , 2 mM dNTPs, 100 mM Tris-HCl, pH 8.3, 50 mM KC1, 1% Tween 20.
  • Tag DNA polymerase was from Cetus Inc., USA.
  • the thermal cycler was a Perkin-Elmer 9600.
  • the synthetic 58 residue SPA analogue Z [Nilsson et al, Prot.Eng] was subjected to a mutagenesis approach to construct new variants with an altered pi, in order to produce fusion partners for recombinant proteins to be pu- rified by ion-exchange chromatography.
  • the PCR mix (50 ⁇ l ) contained one pmole each of PCR primers ZLIB-3 and ZLIB-5, 5 ⁇ l each of the ligation mix, 10 x PCR buffer and 10 x CHASE, 1 unit of Tag polymerase and sterile water to 50 ⁇ l.
  • the temperature cycling programme was: 96°C, 1 min, 60°C, 1 min and 72°C, 2 min, repeated for 35 cycles.
  • Ana ⁇ lysis by 1% agarose gel electrophoresis showed a band of the expected size of 179 bp, showing the feasibility of the assembly concept.
  • the 179 bp band from the PCR of the Z(Acid)-library was cut out from the gel and purified (GenecleanTM, Bio 101, Inc.
  • the temperature stability of the Z conformation was determined by following the ellipticity at 222 run by cir ⁇ cular dichroism (CD) spectroscopy through a temperature scan. This wavelength is used to monitor the presence of ⁇ -helicity of Z [Cedergren et al. 1993 Prot. Eng. 6:441- 448] .
  • the experiment was performed at a rather low pH (approximately 2.9) in order to destabilize the molecule since the mid-point of temperature denaturation (Tm) is ⁇ 95°C at neutral pH (data not shown), which is outside the range that can be determined by a complete scan through the transition under normal atmospheric pressure.
  • the experiment shows (Fig. 4) that the Tm (as defined by the inflexion point of the temperature scan) of the Z domain is as high as 71°C at pH 2.9. This demonstrates the ex- treme temperature stability of the ⁇ -helices of the Z molecule.
  • the experiment was performed in a J-720 spectro- polarimeter (JASCO, Japan) and the temperature was con ⁇ trolled by circulating water through the cuvette holder from a NESLAB water bath. The temperature was monitored in the cuvette through a micro sensor device (JASCO, Japan).
  • the buffer was 50 mM acetic acid, pH 2.9.
  • the protein was domain Z [Cedergren et al 1993 Prot. Eng. 6:441-448] at a protein concentration of 50 ⁇ g/mL and the cuvette cell path length was 1 cm.
  • the temperature scan speed in the experiment was 50°C/h. 23
  • Example 3 Characterization of proteins derived from the acid Z-library.
  • the restricted fragments (approximately 5 ⁇ g) were purified by phenol/chloroform/isoamylaJcohol extraction followed by additional washing with chloroform and later ethanol precipitated before ligation at 15 C C over night to Mlu I-Nhe I cleaved pKNl vector (1 ⁇ g) (see below) using 13.5 Weiss units of T4 DNA ligase.
  • the ligation mixture was heat-treated at 70°C for 20 min, extracted with phenol/chloroform/isoamylalcohol followed by washing with chloroform, ethanol precipitated and redissolved in 20 ⁇ l of sterile water.
  • the phagemid vector pK l (figure 9) was constructed in several steps as follows.
  • a double stranded linker encoding the invariant residues 44-58 of the Z-domain was formed from oligonucleotides ZLIB-6 and ZLIB-7 and cloned as a Mlu l-Xho I fragment into phagemid pKP9S6 (A kind gift from Dr. Lars Abrahmsen. Pharmacia BioScience Center, Sweden),- resulting in pK .
  • Plasmid pKP9S6 encodes the E. coli Omp A leader peptide followed by residues 249-406 of fd filamentous phage coat protein ' 3 (Lowman et al.
  • a gene fragment encoding a monovalent serum albumin binding region derived from streptococcal protein G was amplified by PCR from the plasmid pB2T (Eliasson et al, Molecular Immunol, 28, 1055- 1061), using primers ABP- 1 and ABP-2 (which contain Xn' o I and Sal I recognition sites, respectively) and cloned into Xho I restricted plasmid pKN, yielding pKNl.
  • This phagemid vector thus encodes for the Omp A signal peptide, the third helix of the wild type Z domain followed by a 46 residue albumin binding protein (ABP) linked to residues 249-406 of fd phage protein III and is adapted for inse ⁇ ion of ⁇ sp 3l/Nhe I-digested PCR products encoding variegated helices one and two of the Z domain.
  • ABC albumin binding protein
  • the solid-phase DNA sequencing of the PCR products was performed employing the FITC labeled sequencing primers NOKA-3 (for the immobilized strand) and ABP-2 (for the eluted strand) on a robotic workstation (BiomekTM 1000, Beckman Instruments, Fullerton, CA) and an Automated Laser Fluorescent (A.L.F.) DNA SequencerTM (Pharmacia Biotech, Sweden) as described by Hultman and coworkers (Hultman et al, ( 19S9) Nucleic acids Research, 17, 4937-4946).
  • the periplasmic content from the cells was subjected to affinity chromatography on HSA-Sepharose as described by Nygren and coworkers (Nygren et al, (1988) J. Mol. Recognit., 1, 69-74) and analyzed by SDS/PAGE on a homogeneous 12% slab gel (BioRad Inc., USA), which was stained with Coomassie Brilliant Blue R-250.
  • SDS- PAGE analysis Figure 10 of purified proteins suggest that the acid Z variants analyzed are stably expressed in E. coli.
  • the proteins Z(wild 5 type) and the acid variants no. 10 and no. 12 were each (5 ⁇ g) dissolved in 300 ⁇ l of 20 mM Piperazine buffer (pH 5.5) and separately applied at 100 ⁇ l/min on a MonoQ. PC 1.6/5 column (Pharmacia, Sweden). Elution of the proteins were performed by applying a NaCl gradient in Piperazine buffer (pH 5.5) (Sigma, USA) ranging from 0-50 % NaCl in 20 min.
  • the series of experiments performed on the two acid Z-variant proteins shows that the expression behaviour, proteolytic stability and secondary structure content of the variants were unchanged when compared to the native Z-domain. Furthermore, a novel functions were introduced into the two Z-variants by the substitution of surface located positions with acid amino acids.
  • the two acid variants can be used e.g. as 0 fusion partners to facilitate purification of recombinant proteins by ion exchange chromatography at low pH. Thus, it is showed that among the members of the acid Z- library, variants with novel functions can be isolated.
  • a library of Z-variants was assembled using a solid-phase gene assembly strategy (see example 1). Most of the amino acid residues suggested to take part in the binding Q to Fc (Deisenhofer, (1981) Biochemistry, 20. 2361-2370) were found to be on the molecule surface (Q9, Q10, Ni l, F13, Y14, L17, N28, Q32 and K35), and therefore included in the mutagenesis. In addition, based on their surfacial location, other residues (H18, E24, E25 and R27) were also decided to be included. In total, 13 residues in the Z scaffold where thus chosen for simultaneous and random 5 mutagenesis.
  • a set of oligonucleotides ( Figure 6) were synthesized for construction of the library of surface mutants of the 58-residues monovalent IgG-binding domain denoted Z.
  • the chosen NNK degeneracy includes 32 codons covering all 20 amino acids, including the TAG (amber) termination signal.
  • Oligonucleotide ZLIB- 1 was synthesized with a 5' biotin group to enable robust anchoring onto streptavidin-coated paramagnetic beads used as solid support during the gene assembly.
  • This ZLIB- 1 oligonucleotide, together with its complementary sequence (ZLIB-2) encodes residues 1-8 of the Z domain, preceeded by the first six residues of region E of protein A which were included to facilitate the E. coli secretion of the Z variants (Abrahmsen et al, (1986) EMBO J., 4, 3901-3906).
  • the oligonucleotides DEGEN- 1 and DEGEN-2 (Table I) encode the two mutated helices of the Z domain, respectively, normally involved in Fc-binding. Theoretically, full and simultaneous NNK degeneracy at the 13 selected positions would yield a combinatorial library of appr. 8* 10 ⁇ protein variants encoded by 3.7* 10 ⁇ different DNA sequences. However, here the assembly of the library was initiated by the immobilization of appr. 15 pmole of prehybridized oligonucleotides ZLIB-1 and ZLIB-2 ( Figure 6), which limits the theoretical size of the Z-library to appr. 0.9* 10 1 - 5 different DNA sequences encoding appr. 2* 10 ⁇ Z variants.
  • oligonucleotides ZLIB-3 and ZLIB-5 containing the recognition sequences for the endonucleases Esp 3 I and Nhe I respectively, were used as primers for PCR amplification of the assembled constructs using one tenth of t he bead-immobilized ssDNA as template (theoretically corresponding to 2* 10 ⁇ protein variants).
  • oligonucleotides ZLIB-2, BRIDGE and ZLIB-5 were first eluted with alkali. The resulting PCR product was analysed by agarose gel electrophoresis and found to be homogenous and of the expected size. 179 bp.
  • the PCR product was subcloned into the pK l phagemid vector containing the gene for residues 44-58 of the wild type Z domain in frame with a truncated version of the fd phage coat protein 3 gene for surface display on phage particles upon helper phage s perinfection of phagemid transformed E. coli cells (Lowman et al, ( 1991) Biochemistry, 30, 10S32-10S44) ( Figure 9).
  • the phagemid vector contains an interspaced in-frame cassette encoding a 5 kDa (46 aa) serum albumin binding region (denoted .ABP) derived from streptococcal protein G (Nygren et al, (1988) J.
  • the serum albumin binding activity can potentially be used for pre-selection of phage panicles earning recombinant molecules, prior to the panning for Z variants with new binding functions, to decrease the background originating from unspecifically bound non-recombinant phage particles.
  • PCR screening using the oligonucleotides PJT-27 and NOKA- 2 of 25 clones showed that over 959c (24/25) of the clones contained an insert of the expected length, suggesting that the gene assembly procedure was carried out with high efficiency.
  • Fo ⁇ yfive transformants were randomly selected and subjected to direct solid phase DNA sequencing (see Example 3) in order to further analyze the quality and heterogeneity of the library. Approximately 699 of the clones were correct, containing wild type and degenerate codons at expected positions. The remaining clones had spurious discrepancies which in part can be attributed to the oligonucleotide synthesis or errors introduced during PCR.
  • Soluble proteins from the periplasm of IPTG-induced cultures were subjected to HSA-affmity chromatography employing the .ABP-tail for general and efficient recovery (Nygren et al, (19SS) J. Mol. Recognit., 1, 69-74). For all proteins appr. 1.5-2.5 mg/L culture could be recovered, indicating similar production and secretion efficiencies for the variants and the wild type domain.
  • the results from a SDS-PAGE analysis ( Figure 16) of purified proteins suggest that the four Z variants analyzed are stably expressed in E. coli.

Abstract

Novel proteins obtainable by mutagenesis of surface-exposed amino acids of domains of natural bacterial receptors, said proteins being obtained without substantial loss of basic structure and stability of said natural bacterial receptors; proteins which have been selected from a protein library embodying a repertoire of said novel proteins; and methods for the manufacture of artificial bacterial receptor structures.

Description

Bacterial receptor structures
The present invention relates to new bacterial re- =ptor structures originating from natural bacterial re- ceptor structures which have been modified in regard to amino acid residues involved in the original interaction function, whereby said original interaction function has been substantially inhibited and replaced by a modified interaction function directed to a desired interaction partner.
Several bacteria known to invade mammals have evolved surface proteins capable of binding to a variety of sub¬ stances including host specific carbohydrates and prote¬ ins. Several such receptors from Gram-positive bacterial pathogens have been isolated and characterized in detail as will be shown below. Most well-characterized are the Fc receptors, named after the capability of binding to the constant Fc part of IgG. Based on binding experiments to IgG from various mammalian sources, and subclasses there- of, Fc receptors have been divided into six types I-VI.
The receptor- from S.aureu≤, protein A [SPA], defining' he type I receptor, has been the subject of immense studies. SPA binds IgG from most mammalian species, including man. Of the four subclasses of human IgG, SPA binds to IgGl, and IgG4 but shows very weak or no interaction with IgG3 [Eliasson, M. et al, 1989 J.Biol.Chem. 9:4323-4327]. This pseudoimmune reaction has been used for more than 20 years for the purification and detection of antibodies in diagnostic, research and therapeutic applications. Clo- ning, sequencing and EscΛeric a coli expression of defi¬ ned fragments of the SPA gene revealed a highly repetitive organization, with five IgG binding domains [E-D-A-B-C] , a cell wall spanning region and membrane anchoring sequence [XM] [Uhlen, M. et al, 1984 J.Biol.Chem. 259:1695-1702; Moks, T. et al, 1986 Eur.J.Biochem. 156:637-643]. A vast number of plasmid vectors have been constructed, allowing gene fusions to different fragments of the gene for the purpose of fusion protein production in different hosts [Nilsson B. and Abrahmsen, L. 1990 Meth.Enz. 185:144-161] (Fig. 2a).
The structure for a complex between human Fc [IgGl] and a single domain [B] of SPA has been determined by X- ray crystallography at a resolution of 2.8 A [Deisenhofer, J. et al 1981 Biochemistry 20:2361-2370]. Based on this structure and additional information from NMR experiments, the B domain can be viewed as a compact structure consis- ting of three anti-parallel -helices connected with loops. In the Fc binding, which is of both electrostatic and hydrophobic nature, only side chains of residues from helices 1 and 2 are involved, whilst the third helix is not participating in the binding. Based on this domain B, a synthetic IgG-binding domain [Z] [Nilsson, B. et al 1987 Prot.Eng. 1:107-113] has been constructed, suitable as fu¬ sion partner for the production of recombinant proteins which allows purification by IgG affinity chromatography. The high solubility and the stable structure of the Z domain has been utilized for production, purification and renaturation of a large number of recombinant proteins. [Josephsson, S. and Bishop, R. Trends Biotechnol. 6:218- 224; Samuelsson, E. et al 1991 Bio.Technol. 9:363-366]
Streptococcal strains of serological groups C and G display a binding repertoire for mammalian IgGs, including human IgG3, which is even broader than for the type I re¬ ceptor. The name protein G was suggested for this type III receptor from group G streptococci. In 1986 Olsson and co- workers reported on the cloning and sequencing of the gene from the serological group G streptococci [G148] [Guss, B. et al, 1987 EMBO J. 5:1567-1575; Olsson, A. et al, 1987 Eur.J.Biochem. 168:319-324]. In analogy with SPA is SPG a repetitively arranged molecule, comprising an IgG-binding region of three homologous domains [C1,C2,C3], spaced by smaller D-regions (Fig. 2A). Compared to SPA, SPG displays a different binding spectra for imrnunoglobulins from dif¬ ferent species and subclasses thereof. The IgG binding domains of protein G are now widely used as an immuno- logical tool, i.e. in the affinity purification of mono¬ clonal antibodies. Production of subfragments constructed by DNA-technology, have shown that an individual C-region is sufficient for full IgG-binding. Recently, the struc¬ ture for a complex between the Cl-domain from SPG and hu¬ man Fc was determined with X-ray crystallography (Fig. 2B). This shows that SPG binds to the CH2-CH3 interface but at a different site compared to SPA. The binding is mainly of electrostatic nature which is in contrast to the large contribution of hydrophobic forces seen for the SPA- Fc interaction. Moreover, the 3-D structure of Cl differs from the X structure in that it is built up by two β- sheets connected by an α-helix [ ββ-a-ββ] . The residues of Cl which according to the structure are involved in the binding, corresponds to the α-helix, the loop and the following β-sheet.
An additional activity of SPG is the capability to bind serum albumin. The binding strength is species depen- dent, and among the tested samples, SPG binds strongest to serum albumin from rat, man and mouse. Production and bin¬ ding studies of subfragments of SPG shows that the two binding activities are structurally separated and that the serum albumin binding function is located at the repeti- tive A-B region [Nygren et al 1990 Eur.J.Biochem. 193:143- 148] . This region has been used for several biotechnolo- gical purposes. Recombinant proteins have been produced as fusions to the region which enables the purification by affinity chroma ography, where human serum albumin most frequently has been used as immobilized ligand. Proteins found to be proteolytically sensitive have been produced as "dual affinity fusions" in which they are flanked by two different affinity tails derived from SPA and SPG, respectively. Purification schemes employing both the N- and C-terminal are thus possible which ensures the recove¬ ry of an intact target protein [Hammarberg et al 1989 Proc.Natl.Acad.Sciences USA 86:4367-4371], The strong and specific binding to serum albumin has also been used for the In vivo stabilization of therapeutic proteins.
Through complex formation with the very long-lived serum albumin, the receptor is carried in the circulation (macaque apes) with a half-life which is close to the half-life for serum albumin itself. Studies in mice with the for HIV/AIDS therapy interesting, but rapidly cleared T-cell receptor CD4, showed that it was substantially sta¬ bilized when fused to the serum albumin binding region, when compared with an unfused control protein [Nygren et al 1991 Vaccines 91 Cold Spring Harbor Press 363-368] . The slow clearance can probably be explained by the complex formation with serum albumin which circumvents elimination by the liver and excretion in the kidney. In order to determine the minimal extension required for maintained binding to serum albumin, several smaller fragments of the A-B region have been produced and ana¬ lyzed. The smallest fragment so far with serum albumin binding activity is a 46 residue fragment ["B2A3"] compri- sing region A3 flanked by 13 and 9 residues, respectively, from regions B2 and S.
Based on homology and binding studies of other par¬ tial fragments SPG is regarded to be trivalent with regard to binding to serum albumin. Similar to the monovalent IgG-binding domains Z and Cl B2A3 is relatively small and shows high solubility and stability and is therefore a suitable candidate for modification.
Summary of the invention The present invention has for its main purpose to provide new bacterial receptor structures by modifying natural bacterial receptors in regard to their original interaction functions to result in new structures having modified interaction functions. Another object of the invention is to provide arti¬ ficial bacterial receptor structures which are stable and more resistant to various conditions, such as increased temperatures.
Yet another object of the invention is to provide artificial bacterial receptor structures, the interaction functions of which have been modified to direct same to other desired interaction partners.
With these and other objects that will be clear from the following disclosure in mind the invention provides for novel proteins obtainable by mutagenesis of surface- exposed amino acids of domains of natural bacterial re- ceptors said proteins being obtained without substantial loss of basic structure and stability of said natural bacterial receptors. Said proteins have preferably been selected from a protein library embodying a repertoire of said novel proteins. In such novel bacterial receptor structures, at least one amino acid residue involved in the interaction fuction of the or._jinal bacterial receptor has been made subject to substitution by another amino acid residue so as to result in substantial loss of the original interaction capacity with a modified interaction capacity being created, said substitution being made with¬ out substantial loss of basic structure and stability of the original bacterial receptor.
It is preferred that said bacterial structures origi¬ nate from Gram-positive bacteria. Among such bacteria there may be mentioned Staphylococcuε aureus ,
Streptococcus pyogenes [group A] , Streptococcus group C,G,L, bovine group G streptococci, Streptococcus zooepidemicus [group C] , Streptococcus zooepidemicuε S212, Streptococcus pyogenes [group A] , streptococci groups A,C,G, Peptostreptococcus magnus, Streptococcus agalactiae [group B] .
Of special interest are thermophilic bacteria evolved to persist in environments of elevated temperatures. Re¬ ceptors from species like e.g. Bacillus εtearotherταo- philuε , Ther uε aquaticuε , Theirmococcuε litoralis and Pyrococcus have the potential of being naturally excep¬ tionally stable, thus suitable for providing structural frameworks for protein engineering according to the in¬ vention.
It is particularly preferred to use as a starting ma¬ terial for the modification of the interaction function bacterial receptor structures originating from staphylo¬ coccal protein A or streptococcal protein G.
Among preferred receptors there may be mentioned bac¬ terial receptors originating from Fc[IgG]receptor type I, type II, type III, type IV, type V and type VI, fibronec- tin receptor, M protein, plasmin receptor, collagen re¬ ceptor, fibrinogen receptor or protein L [K light chains], protein H [human IgG], protein B [human IgA,Al], protein Arp [human IgA] .
Particularly preferred bacterial receptors originate from the Fc[IgG]receptor type I of staphylococcal protein A or the serum albumin receptor of streptococcal protein G.
In order to maintain stability and the properties of the original bacterial receptor structure it is preferred in accordance with the present invention that the substi¬ tution involving amino acid residues taking part in the interaction function of the original bacterial receptor does not involve more than about 50% of the amino acid residues of the original bacterial receptor. It is parti- cularly preferred that not more than about 25% of the amino acid residues of the original bacterial receptor are made subject to substitution.
In regard to the original bacterial receptor struc¬ tures selected for modification of their interaction func- tions it is particularly preferred to use receptors origi¬ nating from the IgG-binding domains Z, Cl, and the serum albumin binding domains B2A3.
In order to maintain as far as possible the stability and properties of the original receptor structure subject to modification in accordance with the present invention it is preferred that substitution thereof involves not more than substantially all of the amino acid residues taking part in the interaction function of the original bacterial receptor.
In order to obtain favourable properties concerning stability and resistance to various conditions it is pre- ferred that the bacterial receptor according to the pre¬ sent invention is comprised of not more than about 100 amino acid residues. It is known from scientific reports that proteins of a relatively small size are fairly resis¬ tant to increased temperatures and also to low pH and cer- tain chemicals. For details concerning temperature resis¬ tance c.f. the article by Alexander et al. in Biochemistry 1992, 31, pp 3597-3603.
With regard to the modification of the natural bacte¬ rial receptor structure it is preferred to perform the substitution thereof by resorting to a genetic enginee¬ ring, such as site-directed mutagenesis.
With regard to the interaction partner of the modi¬ fied natural bacterial receptor a multitude of substances are conceivable, such as proteins, lipids, carbohydrates and inorganic substances. Among carbohydrates examples are blood group determinants and pathogen specific oligo- saccharides.
In regard to proteins conceivable interaction part¬ ners are IGF-I, IGF-II, hGH, Factor VIII, insulin and apolipoprotein and their respective receptors as interac¬ tion partners. Furthermore, by selecting new receptor variants with specificity for different folding forms of proteins, affinity resins or analytical tools to facili¬ tate the isolation of correctly folded molecules can be produced. Further examples are viral coat proteins, bac¬ terial antigens, biotin and cell markers, such as CD 34 and CD 4.
Although the present invention is applicable to a variety of natural bacterial receptors the following illustration of the invention more in detail will be directed to the use of the IgG-binding domains Z, Cl and B2A3. The concept of the present invention residing in the use of artificial bacterial receptors based on the natural structures of naturally occurring bacterial receptors is associated with several advantages. Thus, the invention makes it possible to use robust and stable, highly soluble and secretion competent receptors. This is in contrast to previous techniques based on the use of polyclonals and monoclonals, such as for diagnostic purposes, which are not very stable in connection with storage, varying condi¬ tions, such as varying temperatures etc. Furthermore, the invention makes it possible to modify natural bacterial receptors to obtain desired interaction capacities for specific purposes.
For the selection of such functional variants in a large repertoire, a powerful selection system must be employed. Recent developments in this field offer alter¬ native methods. One of the most important tools for pro¬ tein engineering that has emerged during the last years is the phage display of proteins. By recombinant DNA tech¬ niques, single phage particles can be prepared which on their surface carries a protein fused to a phage-coat protein. By panning from a large pool of phages bearing different proteins, or variants of a specific protein, specific phage clones can be sorted out, which displays a certain binding characteristic [WO92/20791 to Winter et al] . Since the phage particle contains packed DNA encoding the phage protein components, a coupling between the spe¬ cific variant of the displayed protein and the correspon¬ ding genetic information is obtained. Using this techni-
9 que, typically 10 phage clones can simultaneously be generated and subjected to panning for screening of de¬ sired characteristics. The phage display technique can be used for selection of both small peptides as well as more complicated proteins such as antibodies, receptors and hormones. For selection of proteins which cannot be se- creted, which is a prerequisite for phage display, intra- cellular systems have been developed in which the library of proteins are fused to a repressor protein with affinity for a specific plasmid-borne operator region resulting in a coupling between the specific protein variant and the plasmid that encoded it. An alternative to the phages as bearer of protein libraries would be to use bacterial cells. Recently, display of recombinant proteins on the surface of Staphylococcus xyloεuε based on fusions to the cell-wall anchoring domain was demonstrated, which opens the possibility of display also of repertoires of proteins for affinity selection of specific variants [Hansson, M. et al 1992 J.Bacteriology 174:4239-4245]. Furthermore, by structure modelling using computer graphic simulations, predictions of the binding and function of altered vari¬ ants of a protein can theoretically be done before the construction of the gene encoding the protein. As indicated above the present invention describes the construction of novel proteins based on the mutagene¬ sis of surface exposed amino acids of domains derived from bacterial receptors. These artificial bacterial receptors can be selected for different applications using a phage display system. The benefits from using bacterial recep¬ tors as structural frameworks are several. They have evol¬ ved to express a binding function without disturbing the overall structure. They are naturally highly soluble, robust to unphysiological conditions, such as pH and heat, folding efficient and are in addition secretion competent. The invention finds use in several different areas. The introductory part of the above-identified patent specification WO92/20791 gives an excellent survey on an¬ tibodies and their structure. Reference is particularly made to page 1 thereof.
The bacterial receptors SPA and SPG have been widely used in antibody technology for detection and purification of antibodies from e.g. hybridom supernatants and ascites fluids. However, not all antibodies are recognized by these receptors, depending on species and subclass. For the smaller subfragments of antibodies (Fig. 4), SPA and SPG show a limited binding, and efficient tools for gene- ral purification schemes are lacking. However, from a repertoire of mutant receptors including SPA and SPG, forms displaying a broader affinity for antibodies and subfragments thereof can potentially be selected. The complex structural organization of antibodies has a number of consequences for their use in different appli¬ cations as well for the production of recombinant deriva¬ tives. For use in immunosorbents, the arrangement of sub- units connected by disulphide bonds can lead to a leakage of released heavy and light chains from columns. The re¬ quirement of successful docking of the two subunits con¬ tributing to the antigen binding site makes the production in bacteria of small subfragments with a low association difficult. The folding of the antibody is dependent on the formation of intra- and inter chain disulphidebonds, which are not able to form in the intracellular environment of bacterial cells. High-level intracellular expression sys¬ tems for recombinant antibodies leads to the formation of inclusion bodies, which have to be renatured to obtain biological activity. These limitations make it worthwhile to search for alternatives for use as protein domains cap¬ able of specific binding, to replace antibodies in a vast number of applications.
The CDR regions forming the antigen bidning part of an antibody forms a total surface available for the anti¬ gen of approximately 800 A2 , with typical 10-20 residues from the antibody involved in the binding. Using the structure of the complex determined by X-ray crystallo¬ graphy between an individual domain B of SPA and human fc[IgGI] as a starting point about 15 amino acids of the said domain involved in this binding can be determined or postulated. The binding surface of about 600 A2 is of the same order of magnitude as between an antibody and its antigen. By arbitrary in vitro mutagenesis of these posi- tions simultaneously there is obtained a large library of Z variants with modified functional properties. In view of the fact that the regions of the Z domain constituting the very stabile so called three-helix bundle is maintained in its native form spectra of proteins are generated which could be considered as "artificial antibodies" and which have the expected high solubility and excellent folding properties capable of binding to a large number of new ligands. Fusions of these artificial receptors to constant regions can be constructed to recruite effector functions, such as complement binding or triggering of ADCC ( antibody dependent cellular cytotoxicity) . There are several potential advantages of utilizing the SPA structure [Z] as a starting point for such "arti¬ ficial antibodies" or artificial bacterial receptors. For a period of about 10 years a large number of proteins have been produced as fusions to SPA, where one has utilized the unique properties of the fusion partner in expression, refolding and purification. In these applications the Z domain has been found to be extremely soluble, stable against proteases, easy to produce in large amounts and foldable to a correct structure also intracellularly in Escherichia coli (no cysteins). Immunoglobulins (Ig:s) are substantially tetramers built up from so called β-sheet structures which stabilize the orientation of the antigen- binding loops which in turn consist of continuous peptide sequences. This is to be compared to the monomeric Z do- main built up from so called three-helix bundle consisting of three closely packed α-helix structures, where the Fc- binding amino acids are found discontinuously in the se¬ quence but in the folded protein are positioned on one and the same binding surface. This difference with regard to the structural elements contributing to the formation of the binding surface enables new possible conformations which cannot be obtained in natural antibodies. The abili¬ ty of Z to be folded to the native structure also under conditions prevailing in the site of cytoplasma opens the possibility of using also derivatives thereof clinically. Genes coding for artificial antibodies with for example virus-neutralizing capacity can be distributed to cells through so called gene therapy resulting in interrupting the infection at an early stage.
From structure data for the complex between an indi¬ vidual Ig-binding domain [Cl] of SPG and human Fc the binding surface can be studied. The binding which is es¬ sentially of an electrostatic nature involves side chains from amino acids from the α-helix as well as from the sub¬ sequent β-sheet [#3] . These differences in structure com¬ pared to the Z domain makes it useful also to create a library of Cl variants to investigate whether differences in binding patterns for artificial antibodies can be ob¬ served depending on the different conditions as regards the topology of the binding surface. Repertoires based on the structures of these and other receptors therefore of- fer different possibilities in the creation of artificial forms with novel functions.
When producing recombinant proteins the purification of the product is frequently a major problem. By expres¬ sing the target protein as a fusion to a so called affi- nity tail the hybrid product can effectively and selec¬ tively be recovered from the cell lysate or in certain cases from the culture medium by passage through a column containing an immobilized ligand. Several such gene fusion systems have been described which are based on the inter- action of a certain protein with a ligand. For industrial applications it is often desirable to clean effectively the columns between the runs to satisfy purity require¬ ments by authorities. Depending on the nature of proteins the relatively harsh conditions (NaOH, acids, heat) often used for organic or physical matrises, for example in ion exchange chromatography and gel filtration, can normally not be used. Here the use of new ligands based on stable structures originating from bacterial receptors are of great importance. In this connection the Z domain from SPA is an excellent example since said domain can be subjected to such difficult conditions as a pH of 1 or heating to 80°C without denaturating non-reversibly (see Example 2 below). From the library of for example Z variants inte¬ resting protein products can be selected for use immobi¬ lized on a solid phase for affinity chromatography. These protein ligands are resistant to effective purification conditions and are therefore useful repetitively on a large scale. In traditional immuno affinity chromatography where immobilized monoclonal antibodies are used for the selective purification of a certain product there are problems with leakage from the column of subunits (heavy and light chains) of the antibody since it consists of four polypeptide chains linked by cystein bridges. Since the artificial bacterial receptors consist only of one polypeptide chain this problem will be avoided. One par¬ ticular area of interest is selection for binding to car- bohydrates. Lectins, nature's binders to this large and important group of biomolecules, have been found to be difficult to purify and have limited stability. Since the generation of antibodies against carbohydrates has been found to be quite complicated a selection of new arti- ficial lectins will be of great importance to research, diagnostics and therapy.
In the production of recombinant proteins in bacte¬ rial hosts precipitates of the gene product are frequently formed, so called inclusion bodies. In order to obtain a native structure of the protein this must be subjected to refolding in vitro . A limitation in such process one is often confronted with is the f^ct that a great part of the material precipitates in the procedure which results in low yields. By producing the protein with an extension in the form of either a short hydrofilic peptide or an easily soluble complete domain [Samuelsson, E. et al 1991 Bio/Technol. 9:363-366] substantially higher concentra¬ tions of the protein will be obtained without precipita¬ tion taking place during renaturation. For example the high solubility of the said domain enables the use of in¬ creased solubility of proteins in either refolding from inclusion bodies or in so called reshuffling of disulphide bridges. From libraries of artificial receptors new forms can be selected having improved properties to facilitate and even make refolding of recombinant proteins possible (cis-acting chaperones) . Recently a new unit operation for the purification of recombinant proteins based on ion exchange chromatography in so called expanded bed has been described [Hansson, M. et al 1994 Bio/Technol. in press]. In this connection there is used a difference in isoelectric point between the target protein and the proteins of the host cell for selective enrichment on a positively charged ion exchange matrix. By fusion to the acid Z domain (pi 4.7) the ion exchange step can take place at a pH, at which the majori¬ ty of the contaminants were of the opposite charge compa- red to the fusion protein. By constructing libraries of bacterial receptors where a selection of amino acids have been replaced by the acid amino acids aspartate and gluta- mate also very acid and solubility increasing domains can be produced for use as fusion partners in the production of recombinant proteins.
As previously described affinity systems based on protein ligands are not totally suitable for industrial purposes in view of the harsh conditions required in the cleaning of columns. Therefore, there is a need for fusion partners having specific affinity towards simple and cheap organic ligands. Panning of phage display libraries of different bacterial receptors against such ligands provide novel affinity tails suitable for the use as fusion part¬ ners for the production purification of recombinant pro- teins.
The present invention provides means for producing and selecting proteins with novel functions. According to the invention this is achieved by extensive mutation of defined residues of stable domains of bacterial receptors. Due to the novel functions of the artificial bacterial re¬ ceptors, these can be used as specific binders for thera¬ peutic, diagnostic, biotechnology or in research. 17 Figure 9. Phagemid vector pKNl . The library PCR products encoding the variegated helices 1 and 2 (both the acid and the extensive library) was subclo- ned into the phagemid vector, pKNl, containing the gene for residues 44-58 of the wild type Z domain (essentially helix 3), followed by the gene for a 46 residues serum albumin binding region (ABP) derived from streptococcal protein G linked in frame with a truncated version of the M13 phage coat protein 3 gene. The phagemid contains the origin of replication derived from plasmid pBR322 as well as the intergenic region ( f1 ori ) required for packing into the phage particles.
Figure 10. SDS-PAGE. HSA-affinity purified proteins from the periplasm of Eεcherichia coli cells produ¬ cing the wild type Z domain and two different acid Z-variants as ABP fusion proteins encoded from their respective phagemid vectors were analyzed by SDS/PAGE. M, molecular weight mar¬ ker; lane 1, wild type Z domain; lane 2, clone 10; lane 3, clone 12.
Figure 11. CD-data. Overlay plot of CD spectra obtained for the wild type Z domain and two variants of the Z-protein library. The signals of the pro¬ teins were obtained after subtraction of the CD signal contribution of the ABP tail, present during the analysis.
Figure 12. Ion exchange chromatography. The two acid Z- variant proteins no. 10 and no. 12 together with the wild type Z-domain (produced as ABP fusion proteins) were each subjected to analy¬ sis at pH 5.5, employing an anion exchange chromatography column. Elution of the proteins 18 from the column was obtained by a NaCl gradi¬ ent. Top: acid Z-variant no. 12; middle, acid Z-variant no. 10; bottom, Z (wild type). Note that the wild type Z protein was not retarded on the column at this pH.
Figure 13. Z-domain structure. A main-chain trace repre¬ sentation of the model of the structure of the native Z-domain. The structure of helices one and two are from the co-crystal structure be¬ tween domain B of SPA and Fc (Deisenhofer, (1981) Biochemistry, 20, 2361-2370). The third helix was built based on the secondary structu¬ re assignments from NMR spectroscopy (Gouda et al., (1992) Biochemistry, 31, 9665-9672). Non- hydrogen atoms of side-chains of residues that were mutated in the construction of the combi¬ natorial library are displayed as ball-and- stick models. The display was generated by the program MOLSCRIPT (Kraulis (1991) J.Appl.
Cryst., 24, 946-950).
Figure 14. Amino acid sequences. Result from DNA-sequen- cing of 31 randomly chosen Z-variants from the library. The residues subjected to the mutage¬ nesis are boxed. Horisontal lines indicate nu- cleotide identity with the wild type Z sequence listed at the top. Indicated are the clones that were expressed and characterized as fusion proteins to the ABP-tail.
Figure 15. Aminoacid distribution. Result from the statis¬ tical analysis of the deduced amino acids at the mutated positions. In total, 13 residues from 31 clones (403 codons) were included in the calculation. The ratios between observed and expected frequencies are shown for all 20 Figure 9 Phagemid vector pKNl . The library PCR products encoding the variegated helices 1 and 2 (both the acid and the extensive library) was subclo- ned into the phagemid vector, pKNl, containing the gene for residues 44-58 of the wild type Z domain (essentially helix 3), followed by the gene for a 46 residues serum albumin binding region (ABP) derived from streptococcal protein G linked in frame with a truncated version of the M13 phage coat protein 3 gene. The phagemid contains the origin of replication derived from plasmid pBR322 as well as the intergenic region ( f1 ori ) required for packing into the phage particles.
Figure 10, SDS-PAGE. HSA-affinity purified proteins from the periplasm of Eεcherichia coli cells produ¬ cing the wild type Z domain and two different acid Z-variants as ABP fusion proteins encoded from their respective phagemid vectors were analyzed by SDS/PAGE. M, molecular weight mar¬ ker; lane 1, wild type Z domain; lane 2, clone 10; lane 3, clone 12.
Figure 11, CD-data. Overlay plot of CD spectra obtained for the wild type Z domain and two variants of the Z-protein library. The signals of the pro¬ teins were obtained after subtraction of the CD signal contribution of the ABP tail, present during the analysis.
Figure 12, Ion exchange chromatography. The two acid Z- variant proteins no. 10 and no. 12 together with the wild type Z-domain (produced as ABP fusion proteins) were each subjected to analy¬ sis at pH 5.5, employing an anion exchange chromatography column. Elution of the proteins from the column was obtained by a NaCl gradi¬ ent. Top: acid Z-variant no. 12; middle, acid Z-variant no. 10; bottom, Z (wild type). Note that the wild type Z protein was not retarded on the column at this pH.
Figure 13. Z-domain structure. A main-chain trace repre¬ sentation of the model of the structure of the native Z-domain. The structure of helices one and two are from the co-crystal structure be¬ tween domain B of SPA and Fc (Deisenhofer, (1981) Biochemistry, 20, 2361-2370). The third helix was built based on the secondary structu¬ re assignments from NMR spectroscopy (Gouda et al., (1992) Biochemistry, 31, 9665-9672). Non- hydrogen atoms of side-chains of residues that were mutated in the construction of the combi¬ natorial library are displayed as ball-and- stick models. The display was generated by the program MOLSCRIPT (Kraulis (1991) J.Appl.
Cryst., 24, 946-950).
Figure 14. Amino acid sequences. Result from DNA-sequen- cing of 31 randomly chosen Z-variants from the library. The residues subjected to the mutage¬ nesis are boxed. Horisontal lines indicate nu- cleotide identity with the wild type Z sequence listed at the top. Indicated are the clones that were expressed and characterized as fusion proteins to the ABP-tail.
Figure 15. Aminoacid distribution. Result from the statis¬ tical analysis of the deduced amino acids at the mutated positions. In total, 13 residues from 31 clones (403 codons) were included in the calculation. The ratios between observed and expected frequencies are shown for all 20 amino acids as well for the only termination signal (TAG) included in the NNG/T degeneracy profile.
Figure 16. SDS-PAGE analysis. HSA-affinity purified pro¬ teins from the periplasm of E.coli cells produ¬ cing the wild type Z domain and four different Z-variants as ABP fusion proteins encoded from their respective phagemid vectors were analyzed by SDS/PAGE. Lanes 1-5: Reduced conditions.
Lanes 6 and 7: Non-reduced conditions. Lane 1, wild type Z domain; lane 2, clone 16; lane 3, clone 21; lane 4, clone 22; lane 5, clone 24; M, molecular weight marker; lane 6, clone 16 and lane 7, clone 22.
Figure 17. CD-data. Overlay plot of CD spectra obtained for the wild type Z domain and four variants of the α-helical protein surface library. The sig- nals of the variants were obtained after sub¬ traction of the CD signal contribution of the ABP tail, present during the analysis.
Figure 18. Biosensor assay. An overlay plot of sensorgrams obtained from the BIA-core™ analysis of the wild type Z domain and four different variants (no. 16,21,22,24; Figure 4) fused to the ABP tail. The IgG-binding activities of the diffe¬ rent proteins were analyzed using a sensor chip coated with approx. 5000 RU human polyclonal
IgG and injections of 45 μl pulses at 2 μl/min of 1500 nM solutions of the different proteins. Note that the differences in plateau values of signals during the injections of the variants no. 16,21,22 and 24 is due to divergent dilu¬ tions into the driving buffer. All reagents and DNA constructions are available at the department for Biochemistry and Biotechnology, Royal Institute of Technology, Stockholm, Sweden.
Material
The oligonucleotides (Fig. 6) were purchased from Scandinavian Gene Synthesis (Sweden), and phosphorylated where indicated according to [Maniatis et al (1988) Mole¬ cular cloning. A laboratory manual. Cold Spring Harbor Laboratory Press]. ZLIB-1 was biotinylated in the 5 ' -end enabling immobilization on paramagnetic beads M-280 Strep- tavidin purchased from Dynal A/S (Norway). Washing/binding buffer was 1 M NaCl, 10 mM Tris-HCl, pH 7.5, 1 mM EDTA (ethylenediamine tetraacetic acid). The annealing/ligation buffer was 30 mM Tris-HCl, pH 7.5, 10 mM MgCl2, 0.2 mM ATP, 1 mM 1.4 dithiothreitol (DTT). DNA ligase were from Boehringer Mannheim, Germany. 10 x PCR buffer contained 20 rnM MgCl2, 2 mM dNTPs, 100 mM Tris-HCl, pH 8.3, 50 mM KC1, 1% Tween 20. Tag DNA polymerase was from Cetus Inc., USA. The thermal cycler was a Perkin-Elmer 9600. For the tempe¬ rature/stability scanning a J-720 spectropolarimeter (JASCO, Japan) was used. Eεcherichia coli strain RR1ΔM15 [Ruther, U. (1982) Nucl.Acids Res. 10:5765-5772] prepared for competence [Maniatis et al (1988) Molecular cloning. A laboratory manual. Cold Spring Harbor Laboratory Press] was used as host for the transformation. Agar plates con¬ tained 100 μg/ml of ampicillin.
EXAMPLE 1 Construction of an acid Z-library
The synthetic 58 residue SPA analogue Z [Nilsson et al, Prot.Eng] was subjected to a mutagenesis approach to construct new variants with an altered pi, in order to produce fusion partners for recombinant proteins to be pu- rified by ion-exchange chromatography. Based on the crys¬ tal structure of the complex between the B-domain of SPA and human Fcl [Deisenhofer, J. et al 1981, Biochemistry 20:2361-2370], five residues from the B-domain participa¬ ting in the binding were chosen as targets for mutagene¬ sis. These five codons corresponding to the Z-residues No. 9, 11, 14, 27 and 35 positioned in helices 1 and 2 were altered simultaneously using degenerate oligonucleotides with the triplet sequence G(C/A)(C/A) at these positions resulting in the codons for the amino acids alanine (50%), aspartic acid (25%) and glutamic acid (25%), respectively.
Using a solid phase gene assembly strategy [Stahl et al,
5 Biotechniques 14:424-434] a library of genes encoding 3
(243) acidic variants of the synthetic IgG-binding Z- domain was created (Fig. 5). Twenty icrolitres (200 μg) of paramagnetic streptavidin-coated beads were washed with washing/binding buffer and incubated with 15 pmole of pre- hybridized oligonucleotides ZLIB-1 (biotinylated) and ZLIB-2, for 15 min at RT at a final volume of washing/- binding buffer of 40 μl. After ligation and washing, ap¬ proximately 15 pmole each of the oligonucleotides ACID-1 (degenerated), LONGBRIDGE, and ACID-2 (degenerated) and the preannealed linker pair ZLIB-4/ZLIB-5 were added in a repetitive manner, with intervening washing steps accor¬ ding to Stahl et al [Biotechniques 14:424-434]. After com¬ pleted assembly, the different fragments were ligated for 15 min at 37°C. To amplify the amount of DNA coding for the Z(Acid)-library still immobilized onto the beads, a fraction was withdrawn and subjected to PCR. The PCR mix (50 μl ) contained one pmole each of PCR primers ZLIB-3 and ZLIB-5, 5μl each of the ligation mix, 10 x PCR buffer and 10 x CHASE, 1 unit of Tag polymerase and sterile water to 50 μl. The temperature cycling programme was: 96°C, 1 min, 60°C, 1 min and 72°C, 2 min, repeated for 35 cycles. Ana¬ lysis by 1% agarose gel electrophoresis showed a band of the expected size of 179 bp, showing the feasibility of the assembly concept. The 179 bp band from the PCR of the Z(Acid)-library, was cut out from the gel and purified (Geneclean™, Bio 101, Inc. USA) prior to insertion in a plasmid vector (TA-cloning™ kit, Invitrogen, Inc. USA) suitable for solid phase DNA sequencing [Hultman et al, 1988] . After transformation and spreading on ampicillin containing agarplates two colonies were chosen for analy¬ sis of the obtained sequences. The results (Fig. 6) show that the expected degeneracy was found at the desired positions.
EXAMPLE 2
Measurement of the temperature stability of the Z conformation
The temperature stability of the Z conformation was determined by following the ellipticity at 222 run by cir¬ cular dichroism (CD) spectroscopy through a temperature scan. This wavelength is used to monitor the presence of α-helicity of Z [Cedergren et al. 1993 Prot. Eng. 6:441- 448] . The experiment was performed at a rather low pH (approximately 2.9) in order to destabilize the molecule since the mid-point of temperature denaturation (Tm) is ~95°C at neutral pH (data not shown), which is outside the range that can be determined by a complete scan through the transition under normal atmospheric pressure. The experiment shows (Fig. 4) that the Tm (as defined by the inflexion point of the temperature scan) of the Z domain is as high as 71°C at pH 2.9. This demonstrates the ex- treme temperature stability of the α-helices of the Z molecule.
The experiment was performed in a J-720 spectro- polarimeter (JASCO, Japan) and the temperature was con¬ trolled by circulating water through the cuvette holder from a NESLAB water bath. The temperature was monitored in the cuvette through a micro sensor device (JASCO, Japan). The buffer was 50 mM acetic acid, pH 2.9. The protein was domain Z [Cedergren et al 1993 Prot. Eng. 6:441-448] at a protein concentration of 50 μg/mL and the cuvette cell path length was 1 cm. The temperature scan speed in the experiment was 50°C/h. 23 Example 3. Characterization of proteins derived from the acid Z-library.
Two protein variants derived from the acid Z-library were expressed in Escheήchia coli. Durified and characterized using SDS-PAGE, circular dichroism and ion exchange chromatography. The PCR products from a solid phase gene assembly (see example 1) were restricted with 45 U Esp 31 (Labassco AB, Sweden) and 50 U Nhe I (Pharmacia, Sweden) in 200 μl buffer (33 mM Tris-acetate. pH 7.9. 10 mM Mg- acetate, 66 mM potassium-acetate, 0.5 mM DTT and 0.1 mg/ml BSA). The mix was overlaid with mineral oil and incubated at 37°C over night. The restricted fragments (approximately 5 μg) were purified by phenol/chloroform/isoamylaJcohol extraction followed by additional washing with chloroform and later ethanol precipitated before ligation at 15CC over night to Mlu I-Nhe I cleaved pKNl vector (1 μg) (see below) using 13.5 Weiss units of T4 DNA ligase. The ligation mixture was heat-treated at 70°C for 20 min, extracted with phenol/chloroform/isoamylalcohol followed by washing with chloroform, ethanol precipitated and redissolved in 20 μl of sterile water.
The phagemid vector pK l (figure 9) was constructed in several steps as follows. A double stranded linker encoding the invariant residues 44-58 of the Z-domain was formed from oligonucleotides ZLIB-6 and ZLIB-7 and cloned as a Mlu l-Xho I fragment into phagemid pKP9S6 (A kind gift from Dr. Lars Abrahmsen. Pharmacia BioScience Center, Sweden),- resulting in pK . Plasmid pKP9S6 encodes the E. coli Omp A leader peptide followed by residues 249-406 of fd filamentous phage coat protein' 3 (Lowman et al. (1991) Biochemistry, 30, 10832-10844.) under the control of a lac promoter. A gene fragment encoding a monovalent serum albumin binding region derived from streptococcal protein G was amplified by PCR from the plasmid pB2T (Eliasson et al, Molecular Immunol, 28, 1055- 1061), using primers ABP- 1 and ABP-2 (which contain Xn' o I and Sal I recognition sites, respectively) and cloned into Xho I restricted plasmid pKN, yielding pKNl. This phagemid vector thus encodes for the Omp A signal peptide, the third helix of the wild type Z domain followed by a 46 residue albumin binding protein (ABP) linked to residues 249-406 of fd phage protein III and is adapted for inseπion of Εsp 3l/Nhe I-digested PCR products encoding variegated helices one and two of the Z domain.
Freeze competent E. coli RR1AM15 {supEAA lacYl lacZ ara-\Λ galKl xyl-5 mil- l leuBβ proAl (mrcC-mrr) recλ+ rpsL20 thi- \ lambda' F[lccl<l tecZΛMlS]) (Ruther, (1982) Nucleic Acids Research, 10, 5765-5772) cells were transformed with the ligation mixture according to Maniatis and coworkers (Maniatis et al. (1982) Molecular cloning: A Laboratory Manual, Cold Spring Harbor, Cold Spring Harbor Laboratory Press) and plated on agar plates containing 100 μg/ml ampicillin (Sigma. USA) and 1 glucose. Small amount of cells from randomly picked colonies were separately subjected to two-step PCR amplifications (30 cycles: 96°C, 15 s; 72CC, 2 min) on a GeneAmp PCR System 9600 (Perkin Elmer, USA), using 5 pmoles of primers RJT-27 and NOKA-2 (biotinylated) in 20 mM TAPS (pH 9.3), 2 mM MgC , 50 mM KC1, 0.1% Tween 20, 0.2 mM deoxyribonucleoside triphosphates (dNTPs) and 1.0 U of Tag DNA polymerase (Perkin-Elmer). The solid-phase DNA sequencing of the PCR products was performed employing the FITC labeled sequencing primers NOKA-3 (for the immobilized strand) and ABP-2 (for the eluted strand) on a robotic workstation (Biomek™ 1000, Beckman Instruments, Fullerton, CA) and an Automated Laser Fluorescent (A.L.F.) DNA Sequencer™ (Pharmacia Biotech, Sweden) as described by Hultman and coworkers (Hultman et al, ( 19S9) Nucleic acids Research, 17, 4937-4946).
Two clones with the different encoded acid aminoacid substitutions (bold face) at the positions 9, 1 1, 14, 27 and 35 in the Z-domian according to table 1 were selected for further analysis. The wild type Z domain and the two different acid Z-variant proteins (clones no. 10 and 12) were expressed from their respective phagemid vectors as fusions to the serum albumin binding tail (ABP) and purified by human serum albumin-affinity chromatography.
Table 1. Amino acid substitutions for selected clones in the acid Z-library a.
Clone no. Encoded aminio acid at position no.
9 1 1 14 27 35
w.t. Gin Asn Tyr Arg Lys
10 Glu Asp Asp Ala Glu
12 Glu Asp Asp AJa Ala a, Letters in boldface indicate acid atr.inoacids Colonies of E. coli RR1ΔM15 cells harbouring the corresponding phagemid vectors were used to inoculate 100 ml of Tryptic Soy Broth (Difco), supplemented with ampicillin (100 μg/ml). The cultures were grown at 37CC to an
Figure imgf000027_0001
followed by induction with a final concentration of 1 mM IPTG and incubation at 30CC over night. The cells were harvested by centrifugation at approximately 5000 g for 10 min and peripla≤mic proteins released by osmotic shock. The periplasmic content from the cells was subjected to affinity chromatography on HSA-Sepharose as described by Nygren and coworkers (Nygren et al, (1988) J. Mol. Recognit., 1, 69-74) and analyzed by SDS/PAGE on a homogeneous 12% slab gel (BioRad Inc., USA), which was stained with Coomassie Brilliant Blue R-250. For all proteins appr. 1.5-2.5 mg/L culture could be recovered, indicating similar production and secretion efficiencies for the variants and the wild type domain. In addition, the results from the SDS- PAGE analysis (Figure 10) of purified proteins suggest that the acid Z variants analyzed are stably expressed in E. coli.
To investigate if the secondary structure content of the derivatives was preserved after the surface mutagenesis, a subtractive circular dichroism analysis was performed. IgG- or HSA-affinity chromatography purified proteins Z. Z-.ABP, the acid derivatives no. 10 and 12 fused to the ABP tail as well as the ABP-tail itself were subjected to a 250 to 184 nm (far UV) circular dichroism analysis at room temperature using a J-720 spectropolarimeter instrument (JASCO, Japan). The scanning speed was 10 nm/min. The cell pathlength was 1 mm. Solutions (approximately 0.1 mg/ml) of the different proteins were prepared in 20 mM phosphate buffer pH 6.5, supplemented with 0.05 % Tween 20 (Kebo AB, Sweden). Accurate protein concentrations were determined by amino acid analysis on a Beckman 6300 amino acid analyzer equipped with System Gold data handling system. CD signals for the derivatives were obtained by subtracting the signal obtained for the ABP tail, after adjustments for differences in protein concentrations, followed by normalization for amino acid contents.
A comparison of signals obtained from 250 to 184 nm for the wild type Z domain and the acid variants fused to the ABP-tail was performed after subtraction of the contribution from the ABP tail itself. The result shows that for the two acid Z- derivatives, spectra similar to the wild type Z domain were obtained with a characteristic minimum at 208 nm and an inflexion point at 222 nm (Johnson, 1990) (Figure 11). This suggests that the three helix bundle framework is preserved in these mutants. The two Z- variants, no. 10 and 12, contain four and three introduced acid aminoacids, respectively, compared to the native Z domain. In order to investigate if the introduced acidity was reflected as differences in their isoelectric points, they were subjected to a gradient elution from an anion exchange column. The proteins Z(wild 5 type) and the acid variants no. 10 and no. 12 (all produced as .ABP fusion proteins) were each (5 μg) dissolved in 300 μl of 20 mM Piperazine buffer (pH 5.5) and separately applied at 100 μl/min on a MonoQ. PC 1.6/5 column (Pharmacia, Sweden). Elution of the proteins were performed by applying a NaCl gradient in Piperazine buffer (pH 5.5) (Sigma, USA) ranging from 0-50 % NaCl in 20 min. The results from 0 the analysis (figure 12) shows that the two acid Z. -variant proteins were eluted at different NaCl concentrations suggesting clear differences in isoelectric points. In contrast, at the pH chosen during the experiments, the wild type Z-domain did not interact with the resin, and was therefore seen in the flow-through.
5 Thus, the series of experiments performed on the two acid Z-variant proteins shows that the expression behaviour, proteolytic stability and secondary structure content of the variants were unchanged when compared to the native Z-domain. Furthermore, a novel functions were introduced into the two Z-variants by the substitution of surface located positions with acid amino acids. The two acid variants can be used e.g. as 0 fusion partners to facilitate purification of recombinant proteins by ion exchange chromatography at low pH. Thus, it is showed that among the members of the acid Z- library, variants with novel functions can be isolated.
5 Example 4. Construction and characterization of a combinatorial librarv of Z-variants.
A library of Z-variants was assembled using a solid-phase gene assembly strategy (see example 1). Most of the amino acid residues suggested to take part in the binding Q to Fc (Deisenhofer, (1981) Biochemistry, 20. 2361-2370) were found to be on the molecule surface (Q9, Q10, Ni l, F13, Y14, L17, N28, Q32 and K35), and therefore included in the mutagenesis. In addition, based on their surfacial location, other residues (H18, E24, E25 and R27) were also decided to be included. In total, 13 residues in the Z scaffold where thus chosen for simultaneous and random 5 mutagenesis. A set of oligonucleotides (Figure 6) were synthesized for construction of the library of surface mutants of the 58-residues monovalent IgG-binding domain denoted Z. In this library, the codons for Q9, Q 10, N i l , F13, Y14, L17 and H I S located in the first α-he!ix and E24, E25, R27, N28, Q32 and K35 in the second α- helix of the Z domain (Figure 13) were substituted for degenerate NNK (K=G or T) codons using a solid phase strategy utilizing the single stranded degenerate oligonucleotides for the assembly. The chosen NNK degeneracy includes 32 codons covering all 20 amino acids, including the TAG (amber) termination signal.
Oligonucleotide ZLIB- 1 was synthesized with a 5' biotin group to enable robust anchoring onto streptavidin-coated paramagnetic beads used as solid support during the gene assembly. This ZLIB- 1 oligonucleotide, together with its complementary sequence (ZLIB-2) encodes residues 1-8 of the Z domain, preceeded by the first six residues of region E of protein A which were included to facilitate the E. coli secretion of the Z variants (Abrahmsen et al, (1986) EMBO J., 4, 3901-3906). The oligonucleotides DEGEN- 1 and DEGEN-2 (Table I) encode the two mutated helices of the Z domain, respectively, normally involved in Fc-binding. Theoretically, full and simultaneous NNK degeneracy at the 13 selected positions would yield a combinatorial library of appr. 8* 10^ protein variants encoded by 3.7* 10 ^ different DNA sequences. However, here the assembly of the library was initiated by the immobilization of appr. 15 pmole of prehybridized oligonucleotides ZLIB-1 and ZLIB-2 (Figure 6), which limits the theoretical size of the Z-library to appr. 0.9* 101-5 different DNA sequences encoding appr. 2* 10^ Z variants. The assembly was continued by the addition and ligation of a preformed construct, obtained after ligation of equimolar amounts of oligonucleotides DEGEN- 1 and DEGEN-2, facilitated by the bridging oligonucleotide BRIDGE (Figure 6).
To complete the assembly, a fragment consisting of the prehybridized oligo- nucleotides ZLIB-4 and ZLEB-5 was added to the beads for ligation. This fragment encodes the second loop and the first six residues of the unaltered third helix of the Z domain. After completed assembly, oligonucleotides ZLIB-3 and ZLIB-5, containing the recognition sequences for the endonucleases Esp 3 I and Nhe I respectively, were used as primers for PCR amplification of the assembled constructs using one tenth of the bead-immobilized ssDNA as template (theoretically corresponding to 2* 10^ protein variants). To avoid unwanted interference during the amplification, oligonucleotides ZLIB-2, BRIDGE and ZLIB-5 were first eluted with alkali. The resulting PCR product was analysed by agarose gel electrophoresis and found to be homogenous and of the expected size. 179 bp.
The PCR product was subcloned into the pK l phagemid vector containing the gene for residues 44-58 of the wild type Z domain in frame with a truncated version of the fd phage coat protein 3 gene for surface display on phage particles upon helper phage s perinfection of phagemid transformed E. coli cells (Lowman et al, ( 1991) Biochemistry, 30, 10S32-10S44) (Figure 9). In addition, the phagemid vector contains an interspaced in-frame cassette encoding a 5 kDa (46 aa) serum albumin binding region (denoted .ABP) derived from streptococcal protein G (Nygren et al, (1988) J. Mol. Recognit., 1, 69-74; Nilsson et al, (1994) Eur. J. Biochem., 224, 103-108), enabling efficient affinity purification of produced Z variants devoid of their native Fc-binding activity. Furthermore, the serum albumin binding activity can potentially be used for pre-selection of phage panicles earning recombinant molecules, prior to the panning for Z variants with new binding functions, to decrease the background originating from unspecifically bound non-recombinant phage particles.
After transformation, PCR screening (using the oligonucleotides PJT-27 and NOKA- 2) of 25 clones showed that over 959c (24/25) of the clones contained an insert of the expected length, suggesting that the gene assembly procedure was carried out with high efficiency. Foπyfive transformants were randomly selected and subjected to direct solid phase DNA sequencing (see Example 3) in order to further analyze the quality and heterogeneity of the library. Approximately 699 of the clones were correct, containing wild type and degenerate codons at expected positions. The remaining clones had spurious discrepancies which in part can be attributed to the oligonucleotide synthesis or errors introduced during PCR. The correct clones (31 clones) (Figure 14) were further analyzed for codon representation at the 13 degenerate positions. The distribution of the total 403 resulting deduced amino acids among the 32 codons included in the NNK degeneracy profile shows a close correlation with the expected frequencies for these yet unselected clones (Figure 15). To investigate the expression and stability of the Z-variants, four clones (no. 16, 21, 22, 24: Figure 14) with different degrees of substitution as well as the wild type Z domain were produced as ABP fusions encoded from their respective phagemid vectors. Soluble proteins from the periplasm of IPTG-induced cultures were subjected to HSA-affmity chromatography employing the .ABP-tail for general and efficient recovery (Nygren et al, (19SS) J. Mol. Recognit., 1, 69-74). For all proteins appr. 1.5-2.5 mg/L culture could be recovered, indicating similar production and secretion efficiencies for the variants and the wild type domain. The results from a SDS-PAGE analysis (Figure 16) of purified proteins suggest that the four Z variants analyzed are stably expressed in E. coli. The smaller band with HSA-binding activity, seen with different intensities most probably corresponds to the ABP-tail itself (5 kDa), resulting from proteolytic cleavage between the Z variant and the ABP tail. Interestingly, both Z-variants (no. 16 and 22) with introduced cvsteine residues formed dimers, which could be observed under non-reducing conditions during SDS- PAGE (Figure 13; lanes 6 and 7).
To investigate if the secondary structure content of the derivatives was preserved after the extensive surface mutagenesis. a subtractive circular dichroism analysis was performed (see example 3). A comparison of signals obtained from 250.to 184 nm for the wild type Z domain and the four variants fused to the ABP-tail was performed after subtraction of the contribution from the .ABP tail itself. The result showed that for three of the four derivatives spectra similar to the wild type Z domain were obtained, with a characteristic minimum at 208 nm and an inflexion point at 222 nm (Johnson, (1990) Prot. Struct. Funct.. Genet., 1, 205-224) (Figure 17). This suggests that the three helix bundle framework probably is preserved in these mutants. However, for the fouπh derivative (no. 24), a spectrum was obtained which resembles spectra seen for random coils, indicating a low content of secondary structure elements (Johnson, 1990). This derivative contains a glutamine to proline substitution at position 32 in helix 2, suggesting a destabilization leading to a collapse of the helix bundle framework.
In order to further investigate the four Z-variants, the interaction with polyclonal human IgG (hlgG) (Pharmacia .AB) for wild type Z and four different Z variant clones (no. 16, 21, 22, 24; Figure 14) fused to the ABP tail were compared using biosensor technology (BlAcore ' , Pharmacia Biosensor AB, Sweden). The carboxylated dextran layer of a CM-5 sensor chip was activated using N- hydroxysuccinimide (ΝHS) and N-ethyl-.V-[3-diethylaminopropy]]-carbodiimide (EDC) chemistry according to the manufacturers' recommendations. For immo¬ bilization of hlgG, 20 μl of a 500 nM hlgG solution in 50 mM acetate. pH 4 was injected at a flow rate of 5 μl/min over the activated surface, resulting in the immobilization of approximately 5000 resonance units (RU). Fortyfive-microlitre samples of the five fusion proteins, dissolved to approximate concentrations of 1500 nM in ΝaCl/Hepes (10 mM Hepes, pH 7.4, 150 mM ΝaCl, 3.4 mM EDTA, 0.5% surfactant P-20), were injected in separate experiments at a flow rate of 2 μl/min. After each sample injection, the hlgG surface was regenerated with 20 mM HC1. As expected, only the wild type Z-domain showed any detectable Fc-binding activity (Figure 18). In conclusion, the results show that a library of SPA variants with a substituted surface made up from 13 residues located in the α-helices can be constructed. The high degree of conservation of the overall framework of the native Z-domain suggests that derivatives with novel functions grafted onto a stable and soluble scaffold could be isolated for use as artificial antibodies in biochemistry, immunology and biotechnology.

Claims

CLAIMS<
1. Novel proteins obtainable by mutagenesis of surface-exposed amino acids of domains of natural bacte- rial receptors, said proteins being obtained without sub¬ stantial loss of basic structure and stability of said natural bacterial receptors.
2. Proteins according to claim 1, which have been selected from a protein library embodying a repertoire of said novel proteins.
3. Proteins according to claim 1 or 2, which are fused to a phage-coat protein, and wherein said bacterial receptors preferably originate from Gram-positive bacte¬ ria.
4. Proteins according to claim 3, wherein said bacte¬ rial receptors originate from bacteria selected from Staphylococcuε aureuε , Streptococcuε pyogeneε [group A], Streptococcuε group C,G,L, bovine group G streptococci, Streptococcuε zooepidemicus [group C] , Streptococcuε zooepidemicus S212, Streptococcuε pyogeneε [group A], streptococci groups A,C,G, Peptoεtreptococcuε magnuε , Streptococcuε agalactiae [group B] .
5. Proteins according to claim 4, wherein said bac¬ terial receptors originate from staphylococcal protein A or streptococcal protein G.
6. Proteins according to claim 4 or 5, wherein said bacterial receptors originate from receptors selected from Fc[IgG]receptor type I, type II, type III, type IV, type V and type VI, fibronectin receptor, M protein, plasmin re- ceptor, collagen receptor, fibrinogen receptor or protein L [K light chains] , protein H [human IgG] , protein B [human IgA,Al], protein Arp [human IgA] .
7. Proteins according to claim 5, wherein said recep¬ tor originates from the Fc[IgG]receptor type I of staphy- lococcal protein A or the serum albumin receptor of strep¬ tococcal protein G.
8. Proteins according to claim 7, wherein said recep¬ tor originates from the IgG-binding domains Z, Cl, and the serum albumin binding domains B2A3.
9. Proteins according to any preceding claim, wherein said substitution involves at most about 50% of the amino acid residues of the original bacterial receptor.
10. Proteins according to claim 9, wherein said sub¬ stitution involves at most about 25% of the amino acid re¬ sidues of the original bacterial receptor.
11. Proteins according to any of claims 1 to 7, wherein said substitution involves at most substantially all of the amino acid residues taking part in the interac¬ tion function of the original bacterial receptor.
12. Proteins according to any preceding claim, where- in said substitution has been obtained by site-directed mutagenesis.
13. Proteins according to any preceding claim, where¬ in said substitution is directed to creating specific in¬ teraction capacity vis-a-vis substances selected from pro- teins, lipids, carbohydrates and inorganic substances as interaction partners.
14. Proteins according to claim 13, wherein said sub¬ stances are selected from carbohydrates, such as blood group determinants, and pathogen specific oligosaccharides as interaction partners.
15. Proteins according to claim 13, wherein said sub¬ stances are selected from IGF-I, IGF-II, hGH, Factor VIII, insulin and apolipoprotein and their respective receptors as interaction partners.
16. Proteins according to claim 13, wherein said sub¬ stitution is directed to creating specific interaction capacity vis-a-vis substances selected from viral coat proteins, bacterial antigens, biotin, and cell markers, such as CD 34, CD 4.
17. Proteins according to claim 13, wherein said sub¬ stitution is directed to creating specific interaction capacity vis-a-vis antibody fragments, such as Fv, scFv, Fab, and Fc.
18. Proteins according to claim 13, wherein said sub¬ stitution is directed to creating specific interaction capacity vis-a-vis organic ligands.
19. A method for the manufacture of an artificial bacterial receptor structure comprising the steps: a) subjecting the repertoire of novel receptor struc¬ tures obtained according to claim 1 to a selection proce¬ dure based on a desired interaction function; and b) isolating the selected receptor structure.
20. A method according to claim 19, comprising the steps: al ) preparing, by recombinant DNA techniques, phage particles carrying on their respective surfaces proteins from said repertoire of novel receptor structures and fused to phage-coat proteins; a2) panning from a pool of phage particles resulting from step al ) to select specific phage clones displaying desired binding characteristics; and b) isolating said specific phage clones using inter¬ actions associated with said binding characteristics.
21. A method according to claim 19 for selection of receptor structures associated with protein of a non- secretory nature, comprising the steps: a) preparing, by recombinant DNA techniques, fusion proteins, wherein the proteins of said library are fused to a repressor protein with affinity for a specific plas- mid-borne operator region resulting in interaction between a specific protein variant and a plasmid encoding the same; and b) isolating selected proteins using said interac¬ tion.
22. A method according to claim 19, comprising the steps: al ) preparing, by recombinant DNA techniques, bacte¬ rial cells carrying on their respective surfaces proteins from said repertoire of novel receptor structures and fused to cell-wall anchoring domains functional in said bacterial cells; a2) panning from a pool of bacterial cells resulting from step al ) to select specific bacterial clones display- ing desired binding characteristics; and b) isolating said specific clones using interactions associated with said binding characteristics.
PCT/SE1995/000034 1994-01-14 1995-01-16 Bacterial receptor structures WO1995019374A1 (en)

Priority Applications (8)

Application Number Priority Date Filing Date Title
EP95907175A EP0739353B1 (en) 1994-01-14 1995-01-16 Bacterial receptor structures
JP51898495A JP4089920B2 (en) 1994-01-14 1995-01-16 Bacterial receptor structure
DE69533644T DE69533644T2 (en) 1994-01-14 1995-01-16 RECEPTOR STRUCTURES FROM BACTERIA
AU15487/95A AU696186B2 (en) 1994-01-14 1995-01-16 Bacterial receptor structures
CA002181042A CA2181042C (en) 1994-01-14 1995-01-16 Bacterial receptor structures
DK95907175T DK0739353T3 (en) 1995-01-16 1995-01-16 Bacterial Receptor Structures
AT95907175T ATE279439T1 (en) 1994-01-14 1995-01-16 RECEPTOR STRUCTURES FROM BACTERIA
US08/669,360 US5831012A (en) 1994-01-14 1995-01-16 Bacterial receptor structures

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
SE9400088A SE9400088D0 (en) 1994-01-14 1994-01-14 Bacterial receptor structures
SE9400088-2 1994-01-14

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US09/082,468 Continuation US6534628B1 (en) 1994-01-14 1998-05-21 Bacterial receptor structures

Publications (2)

Publication Number Publication Date
WO1995019374A1 true WO1995019374A1 (en) 1995-07-20
WO1995019374A9 WO1995019374A9 (en) 2006-04-13

Family

ID=20392561

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/SE1995/000034 WO1995019374A1 (en) 1994-01-14 1995-01-16 Bacterial receptor structures

Country Status (12)

Country Link
US (2) US5831012A (en)
EP (1) EP0739353B1 (en)
JP (3) JP4089920B2 (en)
AT (1) ATE279439T1 (en)
AU (1) AU696186B2 (en)
CA (1) CA2181042C (en)
DE (1) DE69533644T2 (en)
ES (1) ES2225838T3 (en)
NZ (1) NZ278991A (en)
PT (1) PT739353E (en)
SE (1) SE9400088D0 (en)
WO (1) WO1995019374A1 (en)

Cited By (39)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1997024130A1 (en) * 1995-12-27 1997-07-10 Ngk Insulators, Ltd. CANCEROUS METASTASIS INHIBITOR COMPRISING SURFACE POLYSACCHARIDE OF STREPTOCOCCUS AGALACTIAE Ia TYPE OR Ib TYPE AS PRINCIPAL COMPONENT
WO2000063243A1 (en) 1999-04-19 2000-10-26 Biovitrum Ab Derivatives of the b or z domain from staphylococcal protein a (spa) interacting with at least one domain of human factor viii
WO2002008263A3 (en) * 2000-07-19 2002-06-13 Got A Gene Ab Modified virus having an altered tropism
US6602977B1 (en) 1999-04-19 2003-08-05 Biovitrum Ab Receptor structures
US6740734B1 (en) * 1994-01-14 2004-05-25 Biovitrum Ab Bacterial receptor structures
US6831161B1 (en) 1998-10-21 2004-12-14 Affibody Ab Method of affinity separation and ligands for use therein
EP1600459A3 (en) * 2002-06-28 2005-12-07 Domantis Limited Ligand
WO2007065635A1 (en) * 2005-12-05 2007-06-14 Affibody Ab Polypeptides
EP1826270A1 (en) * 1999-07-20 2007-08-29 Affibody AB Method and library for in vitro selection
EP2014359A1 (en) 2007-07-10 2009-01-14 Millipore Corporation Media for affinity chromatography
WO2009019117A1 (en) * 2007-08-03 2009-02-12 Affibody Ab Igf-1r binding polypeptides and their use
WO2009080811A1 (en) * 2007-12-21 2009-07-02 Affibody Ab Polypeptide libraries with a predetermined scaffold
AU2003217119B2 (en) * 2002-03-25 2010-02-18 Cytiva Bioprocess R&D Ab A mutated immunoglobulin-binding protein
US7993650B2 (en) 2003-07-04 2011-08-09 Affibody Ab Polypeptides having binding affinity for HER2
WO2012083425A1 (en) * 2010-12-21 2012-06-28 The University Of Western Ontario Novel alkali-resistant variants of protein a and their use in affinity chromatography
WO2013022091A1 (en) 2011-08-11 2013-02-14 小野薬品工業株式会社 Therapeutic agent for autoimmune diseases comprising pd-1 agonist
EP2559702A1 (en) 2007-02-08 2013-02-20 Domantis Limited Antibody single variable domains against serum albumin
US8388979B2 (en) 2006-01-31 2013-03-05 Ishihara Sangyo Kaisha, Ltd. Polypeptide having affinity for envelope virus constituent and use thereof in transferring substance into cell
US8501909B2 (en) 2007-12-21 2013-08-06 Affibody Ab Polypeptides having affinity for HER2
WO2013143890A1 (en) 2012-03-28 2013-10-03 Affibody Ab Oral administration
WO2014013016A1 (en) 2012-07-20 2014-01-23 Affibody Ab Method for determining the her2 status of a malignancy
WO2014076179A1 (en) 2012-11-14 2014-05-22 Affibody Ab New polypeptide
US8754196B2 (en) 2011-06-08 2014-06-17 Emd Millipore Corporation Chromatography matrices including novel Staphylococcus aureus protein A based ligands
WO2014103203A1 (en) 2012-12-27 2014-07-03 独立行政法人産業技術総合研究所 Molecule library constructed on the basis of backbone structure of microprotein
US9040661B2 (en) 2010-12-21 2015-05-26 Jsr Corporation Support for affinity chromatography and method for isolating immunoglobulin
US9187535B2 (en) 2007-12-19 2015-11-17 Affibody Ab Polypeptide derived from protein A and able to bind PDGF
US9546198B2 (en) 2007-10-12 2017-01-17 Cancer Research Technology Limited Cyclic peptides as ADAM protease inhibitors
US9920112B2 (en) 2008-08-11 2018-03-20 Emd Millipore Corporation Immunoglobulin-binding proteins with improved specificity
US10072050B2 (en) 2008-12-24 2018-09-11 Emd Millipore Corporation Caustic stable chromatography ligands
WO2018194376A1 (en) 2017-04-18 2018-10-25 앱클론(주) Polypeptide improved in protein purity and affinity for antigen, conjugate thereof with antibody or antigen-binding fragment, and preparation method therefor
US10155792B2 (en) 2012-09-25 2018-12-18 Affibody Ab Albumin binding polypeptide
US10167322B2 (en) 2013-12-20 2019-01-01 Affibody Ab Engineered albumin binding polypeptide
US10329331B2 (en) 2010-07-09 2019-06-25 Affibody Ab Polypeptides
WO2019203600A1 (en) 2018-04-18 2019-10-24 앱클론(주) Switch molecule and switchable chimeric antigen receptor
WO2021006199A1 (en) 2019-07-05 2021-01-14 小野薬品工業株式会社 Treatment of hematologic cancer with pd-1/cd3 dual specificity protein
US10894097B2 (en) 2008-12-22 2021-01-19 General Electric Company HER2 binders
WO2021025140A1 (en) 2019-08-08 2021-02-11 小野薬品工業株式会社 Dual-specific protein
WO2021210939A1 (en) 2020-04-17 2021-10-21 앱클론(주) Anti-her2 affibody, and switchable chimeric antigen receptor using same as switch molecule
WO2022080851A1 (en) 2020-10-13 2022-04-21 인센 주식회사 Testosterone-specific affibody and uses thereof

Families Citing this family (297)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
SE9400088D0 (en) * 1994-01-14 1994-01-14 Kabi Pharmacia Ab Bacterial receptor structures
GB9722131D0 (en) * 1997-10-20 1997-12-17 Medical Res Council Method
SE9704141D0 (en) * 1997-11-12 1997-11-12 Sbl Vaccin Ab New protein and nucleotide sequence, encoding said protein
EP1181052A4 (en) * 1999-05-15 2004-08-11 Univ California San Diego Protein a based binding domains with desirable activities
US7163686B1 (en) 1999-05-15 2007-01-16 The Regents Of The University Of California Protein A based binding domains with desirable activities
CA2390691C (en) * 1999-12-24 2016-05-10 Genentech, Inc. Methods and compositions for prolonging elimination half-times of bioactive compounds
US20040077017A1 (en) * 2001-01-12 2004-04-22 Amelie Karlstrom Detection methods
JP2005504261A (en) * 2001-01-12 2005-02-10 アフィボディ・アーベー Detection method
US20030157561A1 (en) * 2001-11-19 2003-08-21 Kolkman Joost A. Combinatorial libraries of monomer domains
AU2002256371B2 (en) * 2001-04-26 2008-01-10 Amgen Mountain View Inc. Combinatorial libraries of monomer domains
US20050048512A1 (en) * 2001-04-26 2005-03-03 Avidia Research Institute Combinatorial libraries of monomer domains
US20040175756A1 (en) * 2001-04-26 2004-09-09 Avidia Research Institute Methods for using combinatorial libraries of monomer domains
US20050053973A1 (en) * 2001-04-26 2005-03-10 Avidia Research Institute Novel proteins with targeted binding
US20050089932A1 (en) * 2001-04-26 2005-04-28 Avidia Research Institute Novel proteins with targeted binding
US7981863B2 (en) 2001-09-19 2011-07-19 Neuronova Ab Treatment of Parkinson's disease with PDGF
FI115343B (en) * 2001-10-22 2005-04-15 Filtronic Lk Oy Internal multi-band antenna
EP2075256A2 (en) 2002-01-14 2009-07-01 William Herman Multispecific binding molecules
EP1474161A4 (en) * 2002-01-16 2005-06-29 Zyomyx Inc Engineered binding proteins
US20060002935A1 (en) 2002-06-28 2006-01-05 Domantis Limited Tumor Necrosis Factor Receptor 1 antagonists and methods of use therefor
US20050074865A1 (en) 2002-08-27 2005-04-07 Compound Therapeutics, Inc. Adzymes and uses thereof
US20060211044A1 (en) * 2003-02-24 2006-09-21 Green Lawrence R Translucent solid matrix assay device dor microarray analysis
US6861251B2 (en) 2003-02-24 2005-03-01 Pritest, Inc. Translucent solid matrix assay device for microarray analysis
WO2004097368A2 (en) * 2003-04-28 2004-11-11 Ciphergen Biosystems, Inc. Improved immunoassays
ES2639301T3 (en) 2003-04-30 2017-10-26 Universität Zürich Cancer treatment procedures using an immunotoxin
IL157086A0 (en) * 2003-07-24 2004-02-08 Guy Patchornik Multivalent ligand complexes
US7956165B2 (en) 2003-07-24 2011-06-07 Affisink Biotechnology Ltd. Compositions and methods for purifying and crystallizing molecules of interest
US20050181398A1 (en) * 2004-01-16 2005-08-18 Fung Eric T. Specific detection of host response protein clusters
EP1732613A2 (en) 2004-04-06 2006-12-20 Affibody AB Use of serum albumin binding peptides conjugates for the preparation of a medicament
US8092549B2 (en) 2004-09-24 2012-01-10 The Invention Science Fund I, Llc Ciliated stent-like-system
US9011329B2 (en) 2004-04-19 2015-04-21 Searete Llc Lumenally-active device
US8361013B2 (en) 2004-04-19 2013-01-29 The Invention Science Fund I, Llc Telescoping perfusion management system
US8353896B2 (en) 2004-04-19 2013-01-15 The Invention Science Fund I, Llc Controllable release nasal system
US7998060B2 (en) 2004-04-19 2011-08-16 The Invention Science Fund I, Llc Lumen-traveling delivery device
US8019413B2 (en) 2007-03-19 2011-09-13 The Invention Science Fund I, Llc Lumen-traveling biological interface device and method of use
US7850676B2 (en) 2004-04-19 2010-12-14 The Invention Science Fund I, Llc System with a reservoir for perfusion management
US8337482B2 (en) 2004-04-19 2012-12-25 The Invention Science Fund I, Llc System for perfusion management
US7857767B2 (en) 2004-04-19 2010-12-28 Invention Science Fund I, Llc Lumen-traveling device
US9801527B2 (en) 2004-04-19 2017-10-31 Gearbox, Llc Lumen-traveling biological interface device
US8921528B2 (en) * 2004-06-01 2014-12-30 Domantis Limited Bispecific fusion antibodies with enhanced serum half-life
US20060045877A1 (en) * 2004-08-30 2006-03-02 Goldmakher Viktor S Immunoconjugates targeting syndecan-1 expressing cells and use thereof
US7563443B2 (en) * 2004-09-17 2009-07-21 Domantis Limited Monovalent anti-CD40L antibody polypeptides and compositions thereof
KR20070084069A (en) 2004-10-08 2007-08-24 도만티스 리미티드 Single domain antibodies against tnfr1 and methods of use therefor
NZ555464A (en) 2004-12-02 2010-03-26 Domantis Ltd Bispecific domain antibodies targeting serum albumin and glp-1 or pyy
US7741275B2 (en) * 2004-12-22 2010-06-22 Lipopeptide Ab Agents and use thereof
US20070003528A1 (en) 2005-06-29 2007-01-04 Paul Consigny Intracoronary device and method of use thereof
CA2628238A1 (en) 2005-11-07 2007-05-18 The Scripps Research Institute Compositions and methods for controlling tissue factor signaling specificity
US20110183319A1 (en) * 2010-01-22 2011-07-28 Searete Llc, A Limited Liability Corporation Of The State Of Delaware Compositions and methods for therapeutic delivery with microorganisms
US8682619B2 (en) 2005-12-14 2014-03-25 The Invention Science Fund I, Llc Device including altered microorganisms, and methods and systems of use
US20110172826A1 (en) * 2005-12-14 2011-07-14 Amodei Dario G Device including altered microorganisms, and methods and systems of use
US8734823B2 (en) 2005-12-14 2014-05-27 The Invention Science Fund I, Llc Device including altered microorganisms, and methods and systems of use
US8278094B2 (en) 2005-12-14 2012-10-02 The Invention Science Fund I, Llc Bone semi-permeable device
US9220917B2 (en) 2006-04-12 2015-12-29 The Invention Science Fund I, Llc Systems for autofluorescent imaging and target ablation
US20120035438A1 (en) 2006-04-12 2012-02-09 Searete Llc, A Limited Liability Corporation Of The State Of Delaware Path selection by a lumen traveling device in a body tub tree based on previous path
AU2007268364B8 (en) * 2006-05-26 2013-05-16 Obodies Limited OB fold domains
GB0611116D0 (en) 2006-06-06 2006-07-19 Oxford Genome Sciences Uk Ltd Proteins
CN101627055A (en) 2006-09-05 2010-01-13 梅达雷克斯公司 The antibody of bone morphogenetic protein and acceptor thereof and their using method
SI2066351T1 (en) 2006-10-02 2016-02-29 E.R. Squibb & Sons, L.L.C. Human antibodies that bind cxcr4 and uses thereof
US20080096233A1 (en) * 2006-10-20 2008-04-24 Robotti Karla M Isolation Of Immune Complexes
GB0621513D0 (en) 2006-10-30 2006-12-06 Domantis Ltd Novel polypeptides and uses thereof
KR20150067395A (en) 2006-12-01 2015-06-17 메다렉스, 엘.엘.시. Human antibodies that bind cd22 and uses thereof
US7981691B2 (en) * 2006-12-08 2011-07-19 General Electric Company Two helix binders
US7977118B2 (en) * 2006-12-08 2011-07-12 General Electric Company Two helix binders
US8198043B2 (en) * 2006-12-08 2012-06-12 General Electric Company Two helix binders
US7989216B2 (en) * 2006-12-08 2011-08-02 General Electric Company Two helix binders
CL2007003622A1 (en) 2006-12-13 2009-08-07 Medarex Inc Human anti-cd19 monoclonal antibody; composition comprising it; and tumor cell growth inhibition method.
NZ578354A (en) 2006-12-14 2012-01-12 Medarex Inc Antibody-partner molecule conjugates that bind cd70 and uses thereof
EP2121745A2 (en) 2007-02-26 2009-11-25 Oxford Genome Sciences (UK) Limited Proteins
WO2008104803A2 (en) 2007-02-26 2008-09-04 Oxford Genome Sciences (Uk) Limited Proteins
US8303960B2 (en) * 2007-02-27 2012-11-06 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Radiolabeled affibody molecules
WO2008124639A2 (en) * 2007-04-04 2008-10-16 Massachusetts Institute Of Technology Poly (amino acid) targeting moieties
GB0708376D0 (en) 2007-05-01 2007-06-06 Alligator Bioscience Ab Novel polypeptides and uses thereof
BRPI0812400A2 (en) 2007-06-05 2014-10-29 Univ Yale UNIT, HYBRIDOMA, PHARMACEUTICAL COMPOSITION, METHOD FOR IDENTIFYING A UNIT, ANTIBODY ISOLATED TO A SAME ANTIGEN CONNECTION UNIT, PEPTIDES MOLECULE, AND UNIT USE.
WO2009039192A2 (en) 2007-09-17 2009-03-26 The Regents Of The University Of Californina Internalizing human monoclonal antibodies targeting prostate cancer cells in situ
EP2853267B1 (en) 2007-09-21 2016-12-07 The Regents of the University of California Targeted interferon demonstrates potent apoptotic and anti-tumor activities
AR068767A1 (en) 2007-10-12 2009-12-02 Novartis Ag ANTIBODIES AGAINST SCLEROSTIN, COMPOSITIONS AND METHODS OF USE OF THESE ANTIBODIES TO TREAT A PATHOLOGICAL DISORDER MEDIATIONED BY SCLEROSTIN
WO2009070350A1 (en) 2007-11-30 2009-06-04 Siemens Healthcare Diagnostics Inc. Adiponectin receptor fragments and methods of use
RU2537265C2 (en) * 2007-12-26 2014-12-27 Биотест Аг Cd138-targeted cell agents and using them
US9011864B2 (en) * 2007-12-26 2015-04-21 Biotest Ag Method of decreasing cytotoxic side-effects and improving efficacy of immunoconjugates
HUE024291T2 (en) * 2007-12-26 2016-01-28 Biotest Ag Immunoconjugates targeting cd138 and uses thereof
ES2543201T3 (en) * 2007-12-26 2015-08-17 Biotest Ag Methods and agents that improve the direction to tumor cells expressing CD138
WO2009146099A2 (en) 2008-04-02 2009-12-03 Georgia State University Research Foundation, Inc. Contrast agents, methods for preparing contrast agents, and methods of imaging
DK2700651T3 (en) 2008-07-18 2019-07-22 Bristol Myers Squibb Co MONOVALENT COMPOSITIONS FOR CD28 BINDING AND METHODS FOR USING IT
US20100022494A1 (en) * 2008-07-24 2010-01-28 Searete Llc, A Limited Liability Corporation Of The State Of Delaware Method, device, and kit for maintaining physiological levels of steroid hormone in a subject
US20100022497A1 (en) * 2008-07-24 2010-01-28 Searete Llc, A Limited Liability Corporation Of The State Of Delaware Method for treating or preventing a cardiovascular disease or condition utilizing estrogen receptor modulators based on APOE allelic profile of a mammalian subject
US20100061976A1 (en) * 2008-07-24 2010-03-11 Searete Llc, A Limited Liability Corporation Of The State Of Delaware Method for treating or preventing osteoporosis by reducing follicle stimulating hormone to cyclic physiological levels in a mammalian subject
US20100022991A1 (en) * 2008-07-24 2010-01-28 Searete Llc, A Limited Liability Corporation Of The State Of Delaware System and device for maintaining physiological levels of steroid hormone in a subject
ES2752025T3 (en) 2008-07-25 2020-04-02 Wagner Richard W Protein screening methods
SG10201405377XA (en) 2008-08-05 2014-12-30 Novartis Ag Compositions and methods for antibodies targeting complement protein c5
US9187330B2 (en) 2008-09-15 2015-11-17 The Invention Science Fund I, Llc Tubular nanostructure targeted to cell membrane
US20100137787A1 (en) * 2008-12-02 2010-06-03 Searete Llc, A Limited Liability Corporation Of The State Of Delaware Delivery devices for modulating inflammation
US20100135983A1 (en) * 2008-12-02 2010-06-03 Searete Llc, A Limited Liability Corporation Of The State Of Delaware Anti-inflammatory compositions and methods
US20100136097A1 (en) * 2008-12-02 2010-06-03 Searete Llc, A Limited Liability Corporation Of The State Of Delaware Systems for modulating inflammation
US20100136095A1 (en) * 2008-12-02 2010-06-03 Searete Llc, A Limited Liability Corporation Of The State Of Delaware Systems for modulating inflammation
US20100136096A1 (en) * 2008-12-02 2010-06-03 Searete Llc, A Limited Liability Corporation Of The State Of Delaware Systems for modulating inflammation
US20100137246A1 (en) * 2008-12-02 2010-06-03 Searete Llc, A Limited Liability Corporation Of The State Of Delaware Anti-inflammatory compositions and methods
US20100136094A1 (en) * 2008-12-02 2010-06-03 Searete Llc, A Limited Liability Corporation Of The State Of Delaware Systems for modulating inflammation
US20110295090A1 (en) 2008-12-04 2011-12-01 Searete Llc, A Limited Liability Corporation Of The State Of Delaware Systems, devices, and methods including implantable devices with anti-microbial properties
CN102245771B (en) 2008-12-16 2013-04-10 诺瓦提斯公司 Yeast display systems
CA2750581A1 (en) 2009-01-21 2010-07-29 Oxford Biotherapeutics Ltd. Pta089 protein
US8317737B2 (en) * 2009-02-25 2012-11-27 The Invention Science Fund I, Llc Device for actively removing a target component from blood or lymph of a vertebrate subject
US8758324B2 (en) 2010-03-05 2014-06-24 The Invention Science Fund I, Llc Device for actively removing a target cell from blood or lymph of a vertebrate subject
US8246565B2 (en) * 2009-02-25 2012-08-21 The Invention Science Fund I, Llc Device for passively removing a target component from blood or lymph of a vertebrate subject
US8454547B2 (en) 2009-02-25 2013-06-04 The Invention Science Fund I, Llc Device, system, and method for controllably reducing inflammatory mediators in a subject
PT2403878T (en) 2009-03-05 2017-09-01 Squibb & Sons Llc Fully human antibodies specific to cadm1
GB0904355D0 (en) 2009-03-13 2009-04-29 Imp Innovations Ltd Biological materials and uses thereof
US9834815B2 (en) 2009-03-25 2017-12-05 Life Technologies Corporation Discriminatory positive/extraction control DNA
US8728479B2 (en) 2009-03-31 2014-05-20 The Trustees Of The University Of Pennsylvania Antigen-binding proteins comprising recombinant protein scaffolds
KR20120057563A (en) 2009-03-31 2012-06-05 노파르티스 아게 Composition and methods of use for therapeutic antibodies specific for the il-12 receptore betal subunit
EP2414391B1 (en) 2009-04-02 2018-11-28 Roche Glycart AG Multispecific antibodies comprising full length antibodies and single chain fab fragments
NZ595792A (en) 2009-04-20 2014-01-31 Oxford Biotherapeutics Ltd Antibodies specific to cadherin-17
ES2693165T3 (en) 2009-04-23 2018-12-07 Siemens Healthcare Diagnostics Inc. Monomeric and dimeric forms of adiponectin receptor fragments and methods of use
HUE035240T2 (en) 2009-04-27 2018-05-02 Novartis Ag Compositions and methods for increasing muscle growth
US8154285B1 (en) 2009-05-29 2012-04-10 The Invention Science Fund I, Llc Non-external static magnetic field imaging systems, devices, methods, and compositions
US8063636B2 (en) * 2009-05-29 2011-11-22 The Invention Science Fund I, Llc Systems, devices, methods, and compositions including targeted ferromagnetic structures
US8058872B2 (en) 2009-05-29 2011-11-15 The Invention Science Fund I, Llc Systems, devices, methods, and compositions including functionalized ferromagnetic structures
US8106655B2 (en) * 2009-05-29 2012-01-31 The Invention Science Fund I, Llc Multiplex imaging systems, devices, methods, and compositions including ferromagnetic structures
US20100303733A1 (en) * 2009-05-29 2010-12-02 Searete Llc, A Limited Liability Corporation Of The State Of Delaware Systems, devices, methods, and compositions including ferromagnetic structures
IE20090514A1 (en) 2009-07-06 2011-02-16 Opsona Therapeutics Ltd Humanised antibodies and uses therof
DK2464657T3 (en) 2009-08-10 2015-06-29 Morphosys Ag New screening strategies for the identification of antibodies or fragments thereof which bind an antigen with enzymatic activity
US9024766B2 (en) * 2009-08-28 2015-05-05 The Invention Science Fund, Llc Beverage containers with detection capability
US8810417B2 (en) * 2009-08-28 2014-08-19 The Invention Science Fund I, Llc Beverage immersate with detection capability
WO2011029823A1 (en) 2009-09-09 2011-03-17 Novartis Ag Monoclonal antibody reactive with cd63 when expressed at the surface of degranulated mast cells
RU2573915C2 (en) 2009-09-16 2016-01-27 Дженентек, Инк. Protein complexes containing superhelix and/or banding, and their use
JP5954876B2 (en) 2009-10-13 2016-07-20 ナノストリング テクノロジーズ, インコーポレイテッド Protein detection by nanoreporter
EP2470569A1 (en) 2009-10-13 2012-07-04 Oxford Biotherapeutics Ltd. Antibodies against epha10
WO2011054007A1 (en) 2009-11-02 2011-05-05 Oxford Biotherapeutics Ltd. Ror1 as therapeutic and diagnostic target
EP2496944A2 (en) 2009-11-05 2012-09-12 Novartis AG Biomarkers predictive of progression of fibrosis
WO2011080050A2 (en) 2009-12-11 2011-07-07 Novartis Ag Binding molecules
EP2518078B1 (en) * 2009-12-15 2020-05-20 Choe, Muhyeon Method for manufacturing dimers and multimers by increasing the production of bond bridges in a complex of multiple monomers and repeating chains of an affinity domain of a type specifically binding to protein monomers
CN102770767A (en) 2010-02-10 2012-11-07 诺瓦提斯公司 Methods and compounds for muscle growth
AR080793A1 (en) 2010-03-26 2012-05-09 Roche Glycart Ag BISPECIFIC ANTIBODIES
MX346731B (en) 2010-04-23 2017-03-30 Genentech Inc * Production of heteromultimeric proteins.
AU2011249782B2 (en) 2010-05-06 2014-10-02 Novartis Ag Compositions and methods of use for therapeutic low density lipoprotein - related protein 6 (LRP6) multivalent antibodies
ES2659406T3 (en) 2010-05-06 2018-03-15 Novartis Ag Compositions and methods of use for therapeutic antibodies against protein 6 related to low density lipoproteins (LRP6)
US20120100166A1 (en) 2010-07-15 2012-04-26 Zyngenia, Inc. Ang-2 Binding Complexes and Uses Thereof
CA2808185A1 (en) 2010-08-13 2012-02-16 Genentech, Inc. Antibodies to il-1.beta. and il-18, for treatment of disease
CN103080134B (en) 2010-08-20 2015-11-25 诺华股份有限公司 The antibody of EGF-R ELISA 3 (HER3)
WO2012025530A1 (en) 2010-08-24 2012-03-01 F. Hoffmann-La Roche Ag Bispecific antibodies comprising a disulfide stabilized - fv fragment
ES2678145T3 (en) 2010-08-24 2018-08-09 Abbott Laboratories Specific antibodies against HIV core protein and their uses
SG188666A1 (en) 2010-09-30 2013-05-31 Agency Science Tech & Res Methods and reagents for detection and treatment of esophageal metaplasia
EP2625203A1 (en) 2010-10-05 2013-08-14 Novartis AG Anti-il12rbeta1 antibodies and their use in treating autoimmune and inflammatory disorders
WO2012085111A1 (en) 2010-12-23 2012-06-28 F. Hoffmann-La Roche Ag Polypeptide-polynucleotide-complex and its use in targeted effector moiety delivery
US10144950B2 (en) 2011-01-31 2018-12-04 Roche Sequencing Solutions, Inc. Methods of identifying multiple epitopes in cells
WO2016100976A2 (en) 2014-12-19 2016-06-23 Apprise Bio, Inc. Methods for identifying multiple epitopes in selected sub-populations of cells
RU2018108836A (en) 2011-02-04 2019-03-14 Дженентек, Инк. Fc OPTIONS AND METHODS FOR PRODUCING THEM
US10689447B2 (en) 2011-02-04 2020-06-23 Genentech, Inc. Fc variants and methods for their production
WO2012109624A2 (en) 2011-02-11 2012-08-16 Zyngenia, Inc. Monovalent and multivalent multispecific complexes and uses thereof
GB201114858D0 (en) 2011-08-29 2011-10-12 Nvip Pty Ltd Anti-nerve growth factor antibodies and methods of using the same
EP4026845A1 (en) 2011-05-06 2022-07-13 Zoetis Services LLC Anti-nerve growth factor antibodies and methods of preparing and using the same
CA2834983C (en) 2011-05-06 2020-11-17 Nvip Pty Ltd Anti-nerve growth factor antibodies and methods of preparing and using the same
JP6258194B2 (en) 2011-05-06 2018-01-10 ネックスヴェット オーストラリア プロプライエタリー リミテッド Anti-nerve growth factor antibodies and methods of making and using them
ES2704038T3 (en) 2011-05-24 2019-03-13 Zyngenia Inc Multivalent and monovalent multispecific complexes and their uses
WO2012172495A1 (en) 2011-06-14 2012-12-20 Novartis Ag Compositions and methods for antibodies targeting tem8
SI2726094T1 (en) 2011-06-28 2017-04-26 Oxford Biotherapeutics Ltd Therapeutic and diagnostic target
CA2840537C (en) 2011-06-28 2021-12-14 Oxford Biotherapeutics Ltd. Antibodies to adp-ribosyl cyclase 2
SI2726099T1 (en) 2011-07-01 2018-11-30 Novartis Ag Method for treating metabolic disorders
JP2014526886A (en) 2011-07-15 2014-10-09 モルフォシス・アー・ゲー Antibodies cross-reactive with macrophage migration inhibitory factor (MIF) and D-dopachrome tomerase (D-DT)
CN107266577B (en) 2011-10-11 2022-09-13 弗·哈夫曼-拉罗切有限公司 Improved assembly of bispecific antibodies
CN104080471B (en) 2011-10-14 2018-08-10 诺华股份有限公司 Antibody and method for Wnt approach relevant diseases
EP2784159B1 (en) 2011-11-25 2018-05-09 SNU R&DB Foundation Hepatitis b virus-derived cis-regulatory element and use thereof
JP6243345B2 (en) 2011-12-05 2017-12-06 ノバルティス アーゲー Antibody to epidermal growth factor receptor 3 (HER3)
KR20140103135A (en) 2011-12-05 2014-08-25 노파르티스 아게 Antibodies for epidermal growth factor receptor 3 (her3) directed to domain ii of her3
US10117932B2 (en) 2011-12-08 2018-11-06 Biotest Ag Uses of immunoconjugates targeting CD138
CN106831985A (en) 2011-12-21 2017-06-13 诺华股份有限公司 The composition and method of the P factors are targeted for antibody
BR112014019579A2 (en) 2012-02-10 2019-10-15 Genentech, Inc SINGLE CHAIN ANTIBODY, POLYNUCLEOTIDE, VECTOR, HOST CELL, METHOD OF PRODUCTION OF A SINGLE CHAIN ANTIBODY, HETEROMULTYMER AND METHOD OF PRODUCTION
NZ628625A (en) 2012-02-20 2016-03-31 Swedish Orphan Biovitrum Ab Publ Polypeptides binding to human complement c5
AU2013222334A1 (en) 2012-02-23 2014-09-11 President And Fellows Of Harvard College Modified microbial toxin receptor for delivering agents into cells
CA2871882A1 (en) 2012-06-27 2014-01-03 F. Hoffmann-La Roche Ag Method for making antibody fc-region conjugates comprising at least one binding entity that specifically binds to a target and uses thereof
BR112014032193A2 (en) 2012-06-27 2017-06-27 Hoffmann La Roche bispecific antibody production and combination determination methods, bispecific antibody, formulation and use of bispecific antibody
GB201213652D0 (en) 2012-08-01 2012-09-12 Oxford Biotherapeutics Ltd Therapeutic and diagnostic target
WO2014026032A2 (en) 2012-08-08 2014-02-13 Apprise Bio, Inc. Increasing dynamic range for identifying multiple epitopes in cells
JOP20200308A1 (en) 2012-09-07 2017-06-16 Novartis Ag IL-18 binding molecules
WO2014084859A1 (en) 2012-11-30 2014-06-05 Novartis Ag Molecules and methods for modulating tmem16a activities
CN104968678B (en) 2012-12-05 2018-12-11 诺华股份有限公司 Target the composition and method of the antibody of EPO
WO2014093417A1 (en) 2012-12-12 2014-06-19 University Of Virginia Patent Foundation Compositions and methods for regulating erythropoiesis
WO2014120916A1 (en) 2013-02-01 2014-08-07 Bristol-Myers Squibb Company Pegylated domain antibodies monovalent for cd28 binding and methods of use
WO2015198217A2 (en) 2013-02-08 2015-12-30 Novartis Ag Compositions and methods for long-acting antibodies targeting il-17
PL2953969T3 (en) 2013-02-08 2020-02-28 Novartis Ag Anti-il-17a antibodies and their use in treating autoimmune and inflammatory disorders
GB201302447D0 (en) 2013-02-12 2013-03-27 Oxford Biotherapeutics Ltd Therapeutic and diagnostic target
DK2962100T3 (en) 2013-02-28 2021-11-01 Caprion Proteomics Inc TUBERCULOSEBIOMARKEARS AND USES THEREOF
WO2014141064A1 (en) 2013-03-13 2014-09-18 Novartis Ag Notch2 binding molecules for treating respiratory diseases
EP2970479B1 (en) 2013-03-14 2019-04-24 Novartis AG Antibodies against notch 3
BR112015023752B1 (en) 2013-03-15 2023-11-14 Zyngenia, Inc. MODULAR RECOGNITION DOMAIN (MRD), COMPLEX COMPRISING MRD AND CETUXIMAB, USES OF THE COMPLEX TO INHIBIT ANGIOGENESIS AND TREAT CANCER AND PHARMACEUTICAL COMPOSITION COMPRISING SAID COMPLEX
WO2014145252A2 (en) 2013-03-15 2014-09-18 Milone Michael C Targeting cytotoxic cells with chimeric receptors for adoptive immunotherapy
GB201311031D0 (en) * 2013-06-20 2013-08-07 Queen Mary & Westfield College Method
AR096601A1 (en) 2013-06-21 2016-01-20 Novartis Ag ANTIBODIES OF LEXINED OXIDATED LDL RECEIVER 1 AND METHODS OF USE
US9562101B2 (en) 2013-06-21 2017-02-07 Novartis Ag Lectin-like oxidized LDL receptor 1 antibodies and methods of use
US10208125B2 (en) 2013-07-15 2019-02-19 University of Pittsburgh—of the Commonwealth System of Higher Education Anti-mucin 1 binding agents and uses thereof
MX371455B (en) 2013-08-02 2020-01-28 Pfizer Anti-cxcr4 antibodies and antibody-drug conjugates.
TW201536318A (en) 2013-08-14 2015-10-01 Novartis Ag Methods of treating sporadic inclusion body myositis
US10203327B2 (en) 2013-11-05 2019-02-12 Novartis Ag Organic compounds
EP3087101A4 (en) 2013-12-20 2017-12-06 Novartis AG Regulatable chimeric antigen receptor
EP3811970A1 (en) 2014-03-15 2021-04-28 Novartis AG Regulatable chimeric antigen receptor
TW201622746A (en) 2014-04-24 2016-07-01 諾華公司 Methods of improving or accelerating physical recovery after surgery for hip fracture
KR20230164192A (en) 2014-05-06 2023-12-01 제넨테크, 인크. Production of heteromultimeric proteins using mammalian cells
WO2016014553A1 (en) 2014-07-21 2016-01-28 Novartis Ag Sortase synthesized chimeric antigen receptors
EP3660042B1 (en) 2014-07-31 2023-01-11 Novartis AG Subset-optimized chimeric antigen receptor-containing t-cells
JP6649358B2 (en) 2014-08-07 2020-02-19 ノバルティス アーゲー Angiopoietin-like 4 antibodies and methods of use
US9988443B2 (en) 2014-08-07 2018-06-05 Novartis Ag Angiopoetin-like 4 (ANGPTL4) antibodies and methods of use
ES2891332T3 (en) 2014-09-17 2022-01-27 Novartis Ag Targeting cytotoxic cells with chimeric receptors for adoptive immunotherapy
MA41044A (en) 2014-10-08 2017-08-15 Novartis Ag COMPOSITIONS AND METHODS OF USE FOR INCREASED IMMUNE RESPONSE AND CANCER TREATMENT
US11566082B2 (en) 2014-11-17 2023-01-31 Cytiva Bioprocess R&D Ab Mutated immunoglobulin-binding polypeptides
WO2016087416A1 (en) 2014-12-03 2016-06-09 F. Hoffmann-La Roche Ag Multispecific antibodies
UY36449A (en) 2014-12-19 2016-07-29 Novartis Ag COMPOSITIONS AND METHODS FOR ANTIBODIES DIRECTED TO BMP6
AU2015371265B2 (en) 2014-12-24 2021-06-03 Neximmune, Inc Nanoparticle compositions and methods for immunotherapy
EP3280432B8 (en) 2015-04-06 2021-04-14 Subdomain, LLC De novo binding domain containing polypeptides and uses thereof
HUE059218T2 (en) 2015-04-08 2022-11-28 Novartis Ag Cd20 therapies, cd22 therapies, and combination therapies with a cd19 chimeric antigen receptor (car) - expressing cell
AU2016273028B2 (en) 2015-06-05 2019-02-14 Novartis Ag Antibodies targeting bone morphogenetic protein 9 (BMP9) and methods therefor
JOP20200312A1 (en) 2015-06-26 2017-06-16 Novartis Ag Factor xi antibodies and methods of use
CA2993009A1 (en) 2015-07-31 2017-02-09 Research Institute At Nationwide Children's Hospital Peptides and antibodies for the removal of biofilms
US11236159B2 (en) 2015-08-03 2022-02-01 Novartis Ag Methods of treating FGF21-associated disorders
US10000561B2 (en) 2015-09-09 2018-06-19 Novartis Ag Thymic stromal lymphopoietin (TSLP)-binding molecules and methods of using the molecules
CA2996635A1 (en) 2015-09-09 2017-03-16 Novartis Ag Thymic stromal lymphopoietin (tslp)-binding molecules and methods of using the molecules
WO2017066719A2 (en) 2015-10-14 2017-04-20 Research Institute At Nationwide Children's Hospital Hu specific interfering agents
CA3003969A1 (en) 2015-11-06 2017-05-11 Orionis Biosciences Nv Bi-functional chimeric proteins and uses thereof
BR112018010052A2 (en) 2015-11-19 2019-02-05 Asclepix Therapeutics Llc peptides with antiangiogenic, anti-lymphogenic and antiedemic properties and nanoparticle formulations
US10946106B2 (en) 2015-11-30 2021-03-16 The Regents Of The University Of California Tumor-specific payload delivery and immune activation using a human antibody targeting a highly specific tumor cell surface antigen
JP2019506844A (en) 2015-12-18 2019-03-14 ノバルティス アーゲー Antibodies targeting CD32b and methods of use thereof
LT3402880T (en) 2016-01-15 2024-03-12 Thermo Fisher Scientific Baltics Uab Thermophilic dna polymerase mutants
JP6991979B2 (en) 2016-02-05 2022-03-04 オリオニス バイオサイエンシズ ビーブイ CD8 binding substance
US11248057B2 (en) 2016-03-07 2022-02-15 Vib Vzw CD20 binding single domain antibodies
KR20170108203A (en) 2016-03-16 2017-09-27 주식회사 피플바이오 Method for Detecting Aggregate Form of Aggregate-Forming Polypeptides
CN109071647B (en) 2016-04-27 2022-11-22 诺华股份有限公司 Anti-growth differentiation factor 15 antibody and application thereof
DK3452591T3 (en) 2016-05-02 2023-09-18 Encodia Inc MACROMOLECULAR ANALYSIS USING NUCLEIC ACID CODING
US10889615B2 (en) 2016-05-11 2021-01-12 Cytiva Bioprocess R&D Ab Mutated immunoglobulin-binding polypeptides
US10654887B2 (en) 2016-05-11 2020-05-19 Ge Healthcare Bio-Process R&D Ab Separation matrix
US10703774B2 (en) 2016-09-30 2020-07-07 Ge Healthcare Bioprocess R&D Ab Separation method
CN109071613A (en) 2016-05-11 2018-12-21 通用电气医疗集团生物工艺研发股份公司 Isolation medium
JP7106187B2 (en) 2016-05-11 2022-07-26 サイティバ・バイオプロセス・アールアンドディ・アクチボラグ How to save the separation matrix
US10730908B2 (en) 2016-05-11 2020-08-04 Ge Healthcare Bioprocess R&D Ab Separation method
JP6987424B2 (en) 2016-05-11 2022-01-05 サイティバ・バイオプロセス・アールアンドディ・アクチボラグ How to clean and / or disinfect the isolation matrix
CN109563141A (en) 2016-05-13 2019-04-02 奥里尼斯生物科学公司 To the therapeutic targeting of cellular structures
EP3454887B1 (en) 2016-05-13 2021-01-20 Orionis Biosciences BV Targeted mutant interferon-beta and uses thereof
TW201802121A (en) 2016-05-25 2018-01-16 諾華公司 Reversal binding agents for anti-factor XI/XIa antibodies and uses thereof
WO2017216724A1 (en) 2016-06-15 2017-12-21 Novartis Ag Methods for treating disease using inhibitors of bone morphogenetic protein 6 (bmp6)
EA201990868A1 (en) 2016-10-04 2019-09-30 Асклепикс Терапьютикс, Инк. CONNECTIONS AND METHODS FOR ACTIVATING A Tie2 SIGNAL SYSTEM
CA3040802A1 (en) 2016-10-24 2018-05-03 Orionis Biosciences Nv Targeted mutant interferon-gamma and uses thereof
US20180163270A1 (en) 2016-12-12 2018-06-14 Cepheid Integrated immuno-pcr and nucleic acid analysis in an automated reaction cartridge
JP7139332B2 (en) 2016-12-23 2022-09-20 ノバルティス アーゲー Factor XI Antibodies and Methods of Use
US11564982B2 (en) 2017-01-04 2023-01-31 Research Institute At Nationwide Children's Hospital DNABII vaccines and antibodies with enhanced activity
WO2018144999A1 (en) 2017-02-06 2018-08-09 Orionis Biosciences, Inc. Targeted engineered interferon and uses thereof
MX2019009255A (en) 2017-02-06 2019-11-05 Orionis Biosciences Nv Targeted chimeric proteins and uses thereof.
US11246911B2 (en) 2017-02-07 2022-02-15 Vib Vzw Immune-cell targeted bispecific chimeric proteins and uses thereof
WO2018146594A1 (en) 2017-02-08 2018-08-16 Novartis Ag Fgf21 mimetic antibodies and uses thereof
CN110392697A (en) 2017-03-02 2019-10-29 国家医疗保健研究所 There is the antibody and application thereof of specificity to NECTIN-4
CA3056088A1 (en) 2017-03-15 2018-09-20 Research Institute At Nationwide Children's Hospital Composition and methods for disruption of bacterial biofilms without accompanying inflammation
AU2018240117A1 (en) 2017-03-24 2019-09-19 Beth Israel Deaconess Medical Center, Inc. Methods for preventing and treating heart disease
WO2018229715A1 (en) 2017-06-16 2018-12-20 Novartis Ag Compositions comprising anti-cd32b antibodies and methods of use thereof
EP3645710A1 (en) 2017-06-26 2020-05-06 Thermo Fisher Scientific Baltics Uab Thermophilic dna polymerase mutants
CN110785433A (en) 2017-06-28 2020-02-11 诺华股份有限公司 Method for preventing and treating urinary incontinence
EP3661536A4 (en) 2017-08-03 2021-07-21 Asclepix Therapeutics, Inc. Methods for identifying and preparing pharmaceutical agents for activating tie2 receptor
US20210040205A1 (en) 2017-10-25 2021-02-11 Novartis Ag Antibodies targeting cd32b and methods of use thereof
SG11202003924YA (en) 2017-10-31 2020-05-28 Encodia Inc Kits for analysis using nucleic acid encoding and/or label
US11464803B2 (en) 2017-11-14 2022-10-11 Arcellx, Inc. D-domain containing polypeptides and uses thereof
AU2018369639A1 (en) 2017-11-14 2020-04-23 Arcellx, Inc. D-domain containing polypeptides and uses thereof
CA3082410A1 (en) 2017-11-14 2019-05-23 Arcellx, Inc. Multifunctional immune cell therapies
EP3713965A1 (en) 2017-11-22 2020-09-30 Novartis AG Reversal binding agents for anti-factor xi/xia antibodies and uses thereof
CN111556900A (en) 2017-12-22 2020-08-18 赛默飞世尔科技波罗的海封闭股份公司 Polymerase chain reaction compositions comprising amines
EP3749295A4 (en) 2018-02-05 2022-04-27 Orionis Biosciences, Inc. Fibroblast binding agents and use thereof
UY38247A (en) 2018-05-30 2019-12-31 Novartis Ag ANTIBODIES AGAINST ENTPD2, COMBINATION THERAPIES AND METHODS OF USE OF ANTIBODIES AND COMBINATION THERAPIES
DE202019005887U1 (en) 2018-07-03 2023-06-14 Marengo Therapeutics, Inc. Anti-TCR antibody molecules and uses thereof
EP3626265A1 (en) 2018-09-21 2020-03-25 INSERM (Institut National de la Santé et de la Recherche Médicale) Anti-human cd45rc antibodies and uses thereof
US20210347851A1 (en) 2018-09-28 2021-11-11 Novartis Ag Cd19 chimeric antigen receptor (car) and cd22 car combination therapies
EP3856779A1 (en) 2018-09-28 2021-08-04 Novartis AG Cd22 chimeric antigen receptor (car) therapies
EP3861021A4 (en) 2018-10-05 2022-06-22 Research Institute at Nationwide Children's Hospital Compositions and methods for enzymatic disruption of bacterial biofilms
AR117343A1 (en) 2018-12-18 2021-07-28 Novartis Ag REVERSIBLE BINDING AGENTS FOR ANTI-FACTOR XI / XIa ANTIBODIES AND USES OF THEM
AU2020223293A1 (en) 2019-02-15 2021-08-19 Integral Molecular, Inc. Claudin 6 antibodies and uses thereof
WO2020168024A1 (en) 2019-02-15 2020-08-20 Integral Molecular, Inc. Antibodies comprising a common light chain and uses thereof
AU2020235455A1 (en) 2019-03-08 2021-10-28 Oxford Genetics Limited Method of selecting for antibodies
GB201903233D0 (en) 2019-03-08 2019-04-24 Oxford Genetics Ltd Method of selecting for antibodies
EP3941580A1 (en) 2019-03-22 2022-01-26 Reflexion Pharmaceuticals, Inc. D-peptidic compounds for vegf
EP3941581A2 (en) 2019-03-22 2022-01-26 Reflexion Pharmaceuticals, Inc. Multivalent d-peptidic compounds for target proteins
CA3138367A1 (en) 2019-04-30 2020-11-05 Encodia, Inc. Methods for preparing analytes and related kits
US20220251232A1 (en) 2019-05-20 2022-08-11 INSERM (Institut National de la Santé et de la Recherche Médicale) Novel anti-cd25 antibodies
CA3145385A1 (en) 2019-07-08 2021-01-14 Steven D. Goodman Antibody compositions for disrupting biofilms
TW202124446A (en) 2019-09-18 2021-07-01 瑞士商諾華公司 Combination therapies with entpd2 antibodies
JP2022548881A (en) 2019-09-18 2022-11-22 ノバルティス アーゲー ENTPD2 Antibodies, Combination Therapy and Methods of Using Antibodies and Combination Therapy
CA3155930A1 (en) 2019-09-27 2021-04-01 Starkage Therapeutics Senescent cell-associated antigen-binding domains, antibodies and chimeric antigen receptors comprising the same, and uses thereof
WO2021116119A1 (en) 2019-12-09 2021-06-17 INSERM (Institut National de la Santé et de la Recherche Médicale) Antibodies having specificity to her4 and uses thereof
KR20230004510A (en) 2020-03-20 2023-01-06 인썸(인스티튜트 내셔날 드 라 싼테 에 드 라 리셰르셰메디칼르) Chimeric antigen receptor specific for human CD45RC and uses thereof
GB202004514D0 (en) 2020-03-27 2020-05-13 Inst De Medicina Molecular Joaeo Lobo Antunes Treatment of Immunosuppressive Cancer
JP2023525053A (en) 2020-05-12 2023-06-14 インサーム(インスティテュ ナシオナル ドゥ ラ サンテ エ ドゥ ラ ルシェルシェ メディカル) A new method to treat cutaneous T-cell lymphoma and TFH-derived lymphoma
CN116419747A (en) 2020-08-07 2023-07-11 福蒂斯治疗公司 CD46 targeting immunoconjugates and methods of use thereof
JP2023551353A (en) 2020-10-13 2023-12-08 アヴィタイド エルエルシー Three-helix bundle protein affinity ligand library and its use
JP2023550446A (en) 2020-11-20 2023-12-01 インセルム(インスティチュート ナショナル デ ラ サンテ エ デ ラ リシェルシェ メディカル) Anti-CD25 antibody
JP2024504547A (en) 2020-11-20 2024-02-01 インセルム(インスティチュート ナショナル デ ラ サンテ エ デ ラ リシェルシェ メディカル) Anti-CD25 antibody
WO2022130300A1 (en) 2020-12-16 2022-06-23 Molecular Partners Ag Novel slow-release prodrugs
EP4294542A1 (en) 2021-02-19 2023-12-27 Avitide LLC Aav2 affinity agents
JP2024509890A (en) 2021-03-09 2024-03-05 モレキュラー パートナーズ アクチェンゲゼルシャフト Protease cleavable prodrugs
AU2022231913A1 (en) 2021-03-09 2023-09-28 Molecular Partners Ag Novel darpin based cd33 engagers
CA3211368A1 (en) 2021-03-09 2022-09-15 Molecular Partners Ag Novel darpin based cd123 engagers
WO2022216993A2 (en) 2021-04-08 2022-10-13 Marengo Therapeutics, Inc. Multifuntional molecules binding to tcr and uses thereof
WO2023170296A1 (en) 2022-03-11 2023-09-14 Inserm (Institut National De La Sante Et De La Recherche Medicale) Nucleic acid system to specifically reprogram b and t cells and uses thereof
WO2024003380A1 (en) 2022-06-30 2024-01-04 Icm (Institut Du Cerveau Et De La Moelle Épinière) Vascular endothelial growth factor receptor-1 (vegfr-1) inhibitors for promoting myelination and neuroprotection
WO2024008755A1 (en) 2022-07-04 2024-01-11 Vib Vzw Blood-cerebrospinal fluid barrier crossing antibodies
WO2024052503A1 (en) 2022-09-08 2024-03-14 Institut National de la Santé et de la Recherche Médicale Antibodies having specificity to ltbp2 and uses thereof
WO2024077118A2 (en) 2022-10-06 2024-04-11 Bicara Therapeutics Inc. Multispecific proteins and related methods

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4954618A (en) * 1986-02-14 1990-09-04 Genex Corporation Cloned streptococcal genes encoding protein G and their use to construct recombinant microorganisms to produce protein G
US5084559A (en) * 1987-03-27 1992-01-28 Repligen Corporation Protein a domain mutants

Family Cites Families (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5312901A (en) * 1986-02-14 1994-05-17 Pharmacia Lkb Biotechnology Ab Cloned streptococcal genes encoding protein G and their use to construct recombinant microorganisms to produce protein G
US5229492A (en) * 1986-02-14 1993-07-20 Pharmacia Lkb Biotechnology Ab Cloned streptococcal genes encoding protein G and their use to construct recombinant microorganisms to produce protein G
US4879213A (en) * 1986-12-05 1989-11-07 Scripps Clinic And Research Foundation Synthetic polypeptides and antibodies related to Epstein-Barr virus early antigen-diffuse
US5783415A (en) * 1991-03-29 1998-07-21 Genentech, Inc. Method of producing an IL-8 receptor polypeptide
SE9400088D0 (en) * 1994-01-14 1994-01-14 Kabi Pharmacia Ab Bacterial receptor structures
US5877016A (en) * 1994-03-18 1999-03-02 Genentech, Inc. Human trk receptors and neurotrophic factor inhibitors

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4954618A (en) * 1986-02-14 1990-09-04 Genex Corporation Cloned streptococcal genes encoding protein G and their use to construct recombinant microorganisms to produce protein G
US5084559A (en) * 1987-03-27 1992-01-28 Repligen Corporation Protein a domain mutants

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
PROTEIN ENGINEERING, Volume 6, No. 4, 1993, LENA CEDERGREN et al., "Mutational Analysis of the Interaction Between Staphylococcal Protein A and Human IgG1", page 441 - page 448. *
THE JOURNAL OF BIOLOGICAL CHEMISTRY, Volume 263, No. 9, March 1988, MARGARETA ELIASSON et al., "Chimeric IgG-Binding Receptors Engineered from Staphylococcal Protein A and Streptococcal Protein G", page 4323 - page 4327. *

Cited By (69)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6740734B1 (en) * 1994-01-14 2004-05-25 Biovitrum Ab Bacterial receptor structures
US5939404A (en) * 1995-12-27 1999-08-17 Ngk Insulators, Ltd. Cancer metastasis inhibitor containing a streptococcus agalactiae Ia type or Ib type surface polysaccharide as a main ingredient
WO1997024130A1 (en) * 1995-12-27 1997-07-10 Ngk Insulators, Ltd. CANCEROUS METASTASIS INHIBITOR COMPRISING SURFACE POLYSACCHARIDE OF STREPTOCOCCUS AGALACTIAE Ia TYPE OR Ib TYPE AS PRINCIPAL COMPONENT
US6831161B1 (en) 1998-10-21 2004-12-14 Affibody Ab Method of affinity separation and ligands for use therein
WO2000063243A1 (en) 1999-04-19 2000-10-26 Biovitrum Ab Derivatives of the b or z domain from staphylococcal protein a (spa) interacting with at least one domain of human factor viii
US6602977B1 (en) 1999-04-19 2003-08-05 Biovitrum Ab Receptor structures
EP1826270A1 (en) * 1999-07-20 2007-08-29 Affibody AB Method and library for in vitro selection
US7456008B2 (en) 2000-07-19 2008-11-25 Got-A-Gene Ab Modified virus comprising one or more non-native polypeptides
WO2002008263A3 (en) * 2000-07-19 2002-06-13 Got A Gene Ab Modified virus having an altered tropism
AU2003217119B2 (en) * 2002-03-25 2010-02-18 Cytiva Bioprocess R&D Ab A mutated immunoglobulin-binding protein
EP1600459A3 (en) * 2002-06-28 2005-12-07 Domantis Limited Ligand
US7993650B2 (en) 2003-07-04 2011-08-09 Affibody Ab Polypeptides having binding affinity for HER2
US9107965B2 (en) 2005-12-05 2015-08-18 Affibody Ab Polypeptides
US8598113B2 (en) 2005-12-05 2013-12-03 Affibody Ab Polypeptides
US8247375B2 (en) 2005-12-05 2012-08-21 Affibody Ab Polypeptides
WO2007065635A1 (en) * 2005-12-05 2007-06-14 Affibody Ab Polypeptides
EP2431383A1 (en) * 2005-12-05 2012-03-21 Affibody AB Polypeptides
US8388979B2 (en) 2006-01-31 2013-03-05 Ishihara Sangyo Kaisha, Ltd. Polypeptide having affinity for envelope virus constituent and use thereof in transferring substance into cell
EP2559702A1 (en) 2007-02-08 2013-02-20 Domantis Limited Antibody single variable domains against serum albumin
EP2559704A1 (en) 2007-02-08 2013-02-20 Domantis Limited Antibody single variable domains against serum albumin
EP2559703A1 (en) 2007-02-08 2013-02-20 Domantis Limited Antibody single variable domains against serum albumin
US9587009B2 (en) 2007-07-10 2017-03-07 Emd Millipore Corporation Media for affinity chromatography
US8772018B2 (en) 2007-07-10 2014-07-08 Emd Millipore Corporation Media for affinity chromatography
EP2014359A1 (en) 2007-07-10 2009-01-14 Millipore Corporation Media for affinity chromatography
WO2009019117A1 (en) * 2007-08-03 2009-02-12 Affibody Ab Igf-1r binding polypeptides and their use
US8426557B2 (en) 2007-08-03 2013-04-23 Affibody Ab IGF-1R binding polypeptides and their use
US9546198B2 (en) 2007-10-12 2017-01-17 Cancer Research Technology Limited Cyclic peptides as ADAM protease inhibitors
US10472393B2 (en) 2007-10-12 2019-11-12 Cancer Research Technology Limited Method for inhibiting ADAM proteases with cyclic peptides
US9187535B2 (en) 2007-12-19 2015-11-17 Affibody Ab Polypeptide derived from protein A and able to bind PDGF
CN101983204A (en) * 2007-12-21 2011-03-02 阿菲博迪公司 Polypeptide libraries with a predetermined scaffold
US8501909B2 (en) 2007-12-21 2013-08-06 Affibody Ab Polypeptides having affinity for HER2
US10556933B2 (en) 2007-12-21 2020-02-11 Affibody Ab Polypeptide libraries with a predetermined scaffold
JP2011509073A (en) * 2007-12-21 2011-03-24 アフィボディ・アーベー Polypeptide library having a predetermined scaffold
US9469670B2 (en) 2007-12-21 2016-10-18 Affibody Ab Polypeptide libraries with a predetermined scaffold
EP2077272A1 (en) 2007-12-21 2009-07-08 Affibody AB Polypeptide libraries with a predetermined scaffold
US8883120B2 (en) 2007-12-21 2014-11-11 Affibody Ab Polypeptides having affinity for HER2
WO2009080811A1 (en) * 2007-12-21 2009-07-02 Affibody Ab Polypeptide libraries with a predetermined scaffold
CN101983204B (en) * 2007-12-21 2015-06-03 阿菲博迪公司 Polypeptide libraries with a predetermined scaffold
US9920112B2 (en) 2008-08-11 2018-03-20 Emd Millipore Corporation Immunoglobulin-binding proteins with improved specificity
US10894097B2 (en) 2008-12-22 2021-01-19 General Electric Company HER2 binders
US10072050B2 (en) 2008-12-24 2018-09-11 Emd Millipore Corporation Caustic stable chromatography ligands
US11084851B2 (en) 2008-12-24 2021-08-10 Emd Millipore Corporation Caustic stable chromatography ligands
US10329331B2 (en) 2010-07-09 2019-06-25 Affibody Ab Polypeptides
US9040661B2 (en) 2010-12-21 2015-05-26 Jsr Corporation Support for affinity chromatography and method for isolating immunoglobulin
US9051375B2 (en) 2010-12-21 2015-06-09 The University Of Western Ontario Alkali-resistant variants of protein A and their use in affinity chromatography
WO2012083425A1 (en) * 2010-12-21 2012-06-28 The University Of Western Ontario Novel alkali-resistant variants of protein a and their use in affinity chromatography
CN103403020A (en) * 2010-12-21 2013-11-20 Jsr株式会社 Novel alkali-resistant variants of protein a and their use in affinity chromatography
US11633507B2 (en) 2010-12-22 2023-04-25 General Electric Company HER2 binders
US8754196B2 (en) 2011-06-08 2014-06-17 Emd Millipore Corporation Chromatography matrices including novel Staphylococcus aureus protein A based ligands
US9376474B1 (en) 2011-06-08 2016-06-28 Emd Millipore Corporation Chromatography matrices including novel Staphylococcus aureus protein a based ligands
US9018305B2 (en) 2011-06-08 2015-04-28 Emd Millipore Corporation Chromatography matrices including novel Staphylococcus aureus protein a based ligands
US8895706B2 (en) 2011-06-08 2014-11-25 Emd Millipore Corporation Chromatography matrices including novel Staphylococcus aureus protein A based ligands
US9234010B2 (en) 2011-06-08 2016-01-12 Emd Millipore Corporation Chromatography matrices including novel Staphylococcus aureus protein A based ligands
EP3939613A1 (en) 2011-08-11 2022-01-19 ONO Pharmaceutical Co., Ltd. Therapeutic agent for autoimmune diseases comprising pd-1 agonist
WO2013022091A1 (en) 2011-08-11 2013-02-14 小野薬品工業株式会社 Therapeutic agent for autoimmune diseases comprising pd-1 agonist
WO2013143890A1 (en) 2012-03-28 2013-10-03 Affibody Ab Oral administration
WO2014013016A1 (en) 2012-07-20 2014-01-23 Affibody Ab Method for determining the her2 status of a malignancy
US10155792B2 (en) 2012-09-25 2018-12-18 Affibody Ab Albumin binding polypeptide
WO2014076179A1 (en) 2012-11-14 2014-05-22 Affibody Ab New polypeptide
WO2014103203A1 (en) 2012-12-27 2014-07-03 独立行政法人産業技術総合研究所 Molecule library constructed on the basis of backbone structure of microprotein
US9897611B2 (en) 2012-12-27 2018-02-20 National Institute Of Advanced Industrial Science And Technology Molecule library constructed on the basis of backbone structure of microprotein
EP3489396A1 (en) 2012-12-27 2019-05-29 National Institute of Advanced Industrial Science and Technology Molecule library constructed on the basis of backbone structure of microprotein
US10167322B2 (en) 2013-12-20 2019-01-01 Affibody Ab Engineered albumin binding polypeptide
WO2018194376A1 (en) 2017-04-18 2018-10-25 앱클론(주) Polypeptide improved in protein purity and affinity for antigen, conjugate thereof with antibody or antigen-binding fragment, and preparation method therefor
WO2019203600A1 (en) 2018-04-18 2019-10-24 앱클론(주) Switch molecule and switchable chimeric antigen receptor
WO2021006199A1 (en) 2019-07-05 2021-01-14 小野薬品工業株式会社 Treatment of hematologic cancer with pd-1/cd3 dual specificity protein
WO2021025140A1 (en) 2019-08-08 2021-02-11 小野薬品工業株式会社 Dual-specific protein
WO2021210939A1 (en) 2020-04-17 2021-10-21 앱클론(주) Anti-her2 affibody, and switchable chimeric antigen receptor using same as switch molecule
WO2022080851A1 (en) 2020-10-13 2022-04-21 인센 주식회사 Testosterone-specific affibody and uses thereof

Also Published As

Publication number Publication date
EP0739353B1 (en) 2004-10-13
DE69533644T2 (en) 2005-02-17
CA2181042C (en) 2008-04-15
EP0739353A1 (en) 1996-10-30
US5831012A (en) 1998-11-03
JPH09508016A (en) 1997-08-19
AU1548795A (en) 1995-08-01
ES2225838T3 (en) 2005-03-16
JP2007308509A (en) 2007-11-29
US6534628B1 (en) 2003-03-18
JP2005263810A (en) 2005-09-29
ATE279439T1 (en) 2004-10-15
PT739353E (en) 2005-01-31
JP4089920B2 (en) 2008-05-28
WO1995019374A9 (en) 2006-04-13
AU696186B2 (en) 1998-09-03
JP4373461B2 (en) 2009-11-25
SE9400088D0 (en) 1994-01-14
NZ278991A (en) 1997-04-24
CA2181042A1 (en) 1995-07-20
DE69533644D1 (en) 2004-11-18

Similar Documents

Publication Publication Date Title
EP0739353B1 (en) Bacterial receptor structures
US6740734B1 (en) Bacterial receptor structures
Nord et al. A combinatorial library of an α-helical bacterial receptor domain
US20210162319A1 (en) Mutated Immunoglobulin-Binding Polypeptides
Gunneriusson et al. Surface display of a functional single-chain Fv antibody on staphylococci
US8883692B2 (en) Method for cell surface displaying of target proteins using Bacillus anthracis exosporium
Nord et al. Ligands selected from combinatorial libraries of protein A for use in affinity capture of apolipoprotein A-1M and taq DNA polymerase
Jonsson et al. A protein G-related cell surface protein in Streptococcus zooepidemicus
JP3043407B2 (en) Complete synthetic affinity reagent
Ljungquist et al. Thiol‐directed immobilization of recombinant IgG‐binding receptors
AU729449B2 (en) Materials and methods relating to the attachment and display of substances on cell surfaces
CA2258518A1 (en) Recognition molecules interacting specifically with the active site or cleft of a target molecule
US9238810B2 (en) Polypeptide
Gräslund et al. A novel affinity gene fusion system allowing protein A-based recovery of non-immunoglobulin gene products
US5411732A (en) Preparation of fused proteins, antibodies and processes therefore
WO2004106361A2 (en) Peptides for metal ion affinity chromatogrpahy
Derouiche et al. Binding of colicins A and E1 to purified ToIA domains
US5801037A (en) Expression of signal-peptide-free staphylokinases
Jansson et al. A dual-affinity gene fusion system to express small recombinant proteins in a soluble form: expression and characterization of protein A deletion mutants
Ståhl et al. Strategies for gene fusions
JP2584697B2 (en) Recombinant protein A for IgG purification
CN107043421B (en) Single-domain heavy-chain antibody of anti-c-Myc label
CN106831991B (en) Nano antibody of anti-c-Myc label
CN106946990B (en) Nano antibody aiming at c-Myc label
CN106831990B (en) Nano antibody capable of specifically binding c-Myc label

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AM AT AU BB BG BR BY CA CH CN CZ DE DK EE ES FI GB GE HU JP KE KG KP KR KZ LK LR LT LU LV MD MG MN MW MX NL NO NZ PL PT RO RU SD SE SI SK TJ TT UA US UZ VN

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): KE MW SD SZ AT BE CH DE DK ES FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN ML MR NE SN TD TG

DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
121 Ep: the epo has been informed by wipo that ep was designated in this application
WWE Wipo information: entry into national phase

Ref document number: 1995907175

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 278991

Country of ref document: NZ

WWE Wipo information: entry into national phase

Ref document number: 2181042

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 08669360

Country of ref document: US

WWP Wipo information: published in national office

Ref document number: 1995907175

Country of ref document: EP

REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

WWG Wipo information: grant in national office

Ref document number: 1995907175

Country of ref document: EP

COP Corrected version of pamphlet

Free format text: PAGES 15-16, DESCRIPTION, ADDED