CA2105303A1 - Display phage - Google Patents

Display phage

Info

Publication number
CA2105303A1
CA2105303A1 CA002105303A CA2105303A CA2105303A1 CA 2105303 A1 CA2105303 A1 CA 2105303A1 CA 002105303 A CA002105303 A CA 002105303A CA 2105303 A CA2105303 A CA 2105303A CA 2105303 A1 CA2105303 A1 CA 2105303A1
Authority
CA
Canada
Prior art keywords
phage
protein
coat protein
gene
library
Prior art date
Legal status (The legal status 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 status listed.)
Abandoned
Application number
CA002105303A
Other languages
French (fr)
Inventor
William Markland
Bruce L. Roberts
Robert C. Ladner
Arthur C. Ley
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Protein Engineering Corp
Original Assignee
Individual
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
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=24668274&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=CA2105303(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Priority claimed from US07/664,989 external-priority patent/US5223409A/en
Application filed by Individual filed Critical Individual
Publication of CA2105303A1 publication Critical patent/CA2105303A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/43504Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from invertebrates
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K1/00General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length
    • C07K1/04General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length on carriers
    • C07K1/047Simultaneous synthesis of different peptide species; Peptide libraries
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • 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
    • C07K14/24Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from bacteria from Enterobacteriaceae (F), e.g. Citrobacter, Serratia, Proteus, Providencia, Morganella, Yersinia
    • C07K14/245Escherichia (G)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/43504Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from invertebrates
    • C07K14/43513Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from invertebrates from arachnidae
    • C07K14/43522Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from invertebrates from arachnidae from scorpions
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/43504Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from invertebrates
    • C07K14/43563Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from invertebrates from insects
    • C07K14/43572Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from invertebrates from insects from bees
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/81Protease inhibitors
    • C07K14/8107Endopeptidase (E.C. 3.4.21-99) inhibitors
    • C07K14/811Serine protease (E.C. 3.4.21) inhibitors
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/81Protease inhibitors
    • C07K14/8107Endopeptidase (E.C. 3.4.21-99) inhibitors
    • C07K14/811Serine protease (E.C. 3.4.21) inhibitors
    • C07K14/8114Kunitz type inhibitors
    • 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
    • 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
    • C12N7/00Viruses; Bacteriophages; Compositions thereof; Preparation or purification thereof
    • CCHEMISTRY; METALLURGY
    • C40COMBINATORIAL TECHNOLOGY
    • C40BCOMBINATORIAL CHEMISTRY; LIBRARIES, e.g. CHEMICAL LIBRARIES
    • C40B40/00Libraries per se, e.g. arrays, mixtures
    • C40B40/02Libraries contained in or displayed by microorganisms, e.g. bacteria or animal cells; Libraries contained in or displayed by vectors, e.g. plasmids; Libraries containing only microorganisms or vectors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2800/00Properties of cosmetic compositions or active ingredients thereof or formulation aids used therein and process related aspects
    • A61K2800/40Chemical, physico-chemical or functional or structural properties of particular ingredients
    • A61K2800/57Compounds covalently linked to a(n inert) carrier molecule, e.g. conjugates, pro-fragrances
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/01Fusion polypeptide containing a localisation/targetting motif
    • C07K2319/02Fusion polypeptide containing a localisation/targetting motif containing a signal sequence
    • 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
    • C12N2795/00Bacteriophages
    • C12N2795/00011Details
    • C12N2795/14011Details ssDNA Bacteriophages
    • C12N2795/14022New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes

Abstract

The present invention relates to libraries of display phage which display various mutant epitopic peptides or protein domains with potential to bind to a target material of interest.
Site-specific protease cleavable linkers may be incorporated into these phages to overcome the problem of irreversible binding to target materials. When the displayed insert is a mini-protein, a reagent which cleaves the crosslink may be used to facilitate release of a tightly bound display phage. The invention also relates to providing a more balanced library of epitopic peptide display phage.

Description

W092tl5679 PCT/US92/Ot~39 2t B3~3 IMPROVED EPITOPE DISPLAYING PHAGE

~AC~aROln*D OF ~ Nnn~NTION
Field of the In~ention 5This invention relates to the expression and display of libraries of mu~ated epitopic peptideq or potential binding protein domains on the surface of phage, and the screening of those libraries to identify high affinity species .
Information Disclosure Statement The amino acid sequence of a protein determines its three-dimensional (3D) structure, which in turn determines protein function. Some residues on the polypeptide chain are more important than others in determining the 3D structure of a protein. Substitutions of amino acids that are exposed to solvent are le~s likely to affect the 3D structure than are substitutions at internal loci.
"Protein engineering" is the art of manipulating the sequence of a protein in order to alter its binding characteristics. The factors affecting protein binding are known, but designing new complementary surfaces has proven difficult.
With the development of recombinant DNA technique~, it became possible to obtain a mutant protein by mutating the gene encoding the nat~ve protein and then expressing the mutated gene. Several mutagenesis strategies are known. One, "protein surgery", involves the introduction of one or more predetermined mutations within the gene of choice. A sinqle polypeptide of completely predetermined ~equence is expressed, and its binding characteristics are evaluated.
At the other extreme i~ random mutagenesis by means of rel~tively nonspecific mutagens quch as radiation and Yarious chemical agents.

' , . , ,: , - ~.
. . ~ . .
': ' ' ' ' ' , .

W09~/2~679 PCT/US9~ 3s Q~

It is possible to randomly vary predetermined nucleotides using a mixture of bases in the appropriate cycles of a nucleic acid synthesis procedure. The proportion of bases in the mixture, for each position of a codon, will determine the frequency at which each amino acid will occur in the polypeptides expressed from the degenerate DNA population. Oliphant et al. (OLIP86) and Oliphant and Struhl (OLIP87) have demonstrated ligation and cloning of highly degenerate oligonucleotides, which were used in the mutation of promoters. They suggested that similar methods could be used in the variation of protein coding regions. They do not say how one should:
a) choose protein residues to vary, or b) select or screen mutants with desirable properties. Reidhaar-Olson and Sauer (REID88a) have used synthetic degenerate oligo-nts to ~ary simultaneously two or three residues through all twenty amino acids. See also Vershon et al.
(VERS86a; VERS86b). Reidhaar-Olson and Sauer do not discuss the limits on how many residues could be varied at once nor do they mention the problem of unequal abundance of DNA encoding different amino acids.
A number of researchers have directed unmutated foreign antigenic epitopes to the surface of phage, fused to a native phage surface protein, and demonstrated that the epitopes were recognized by antibodies.
Dulbecco (DULB86) suggest~ a procedure for incor-porating a foreign antigenic epitope into a viral surface protein 80 that the expressed chimeric protein is dis-played on the surface of the virus in a manner ~uch that the foreign epitGpe is accessible to antibody. In 1985 Smith (SMIT85) reported inserting a nonfunctional segment of the EcoRI endonuclease gene into gene III of bacterio-phage fl, n in phase n . The gene III protein is a minor coat protein necessary for infectivity. Smith demons-trated that the recombinant phage were adsorbed byimmobilized antibody raised against the EcoRI endonucle-' .

WO92/15679 PCT/US9'/01~39 2t Q~ ~i3t.

ase, and could be eluted with acid. De la Cruz et al.(DELA88) have expressed a fragment of the repeat region of the circumsporozoite protein from Pla~modium falci~arum on the surface of M13 as an insert in the gene III protein. They showed that the recombinant phage were both antigenic and immunogenic in rabbits, and that such recombinant phage could be used for B epitope mapping.
The researchers suggest that similar recombinant phage could be used for T epitope mapping and ~or vaccine 10 development. -McCafferty ~ al. (MCCA90) expressed a fusion of an Fv fra$ment of an antibody to the N-terminal o~ the pIII
protein. The Fv fragment was not mutated.
Ladner, Glick, and Bird, W088/06630 (publ. 7 Sept.
1988 and having priority from US application 07/021,046, assigned to Genex Corp.) (LGB) speculate that diverse single chain antibody domains (SCAD) may be screened for binding to a particular antigen by varying the DNA
encoding the combining determining regions of a single chain antibody, subcloning the SCAD gene into the gpV
gene of phage lambda 80 that a SCAD/gpV chimera is displayed on the outer surface of phage lambda, and selecting phage which bind to the antigen through affinity chromatography.
Parmley and Smith (PARM88) suggested that an epitope library that exhibits all possible hexapeptides could be constructed and used to isolate epitopes that bind to antibodies. In discu~sing the epitope library, the authors did not suggest that it was desirable to balance the representation of different amino acids. Nor did they teach that the insert should encode a complete domain of the exogenous protein. Epitopes are considered to be unstructured peptides as opposed to structured proteins. Scott and Smith (SCOT90) and Cwirla et al.
(CWIR90) prepared Repitope libraries" in which potential hexapeptide epitopes for a target antibody were randomly .- ,~ . - .
.
.
., '','" ~ .

, WO92/lS679 PCT/US92/01~39 ~ 3;~' mutated by fusing degenerat~ oligonucleQtides, encoding the epitopes, with gene III of fd phage, and expressing the fused gene in phage-infected cells. The cells manufactured fusion phage which displayed the epitopes on their surface; the phage which bound to immobilized antibody were eluted with acid and studied. Devlin et al. ~DEV~90) similarly screened, using M13 phage, for random 15 residue epitopes recognized by streptavidin.
The Scott and Smith, Cwirla et al., and Devlin et ~1~, libraries provided a highly biased sampling of the pos~ible amino acid~ at each position. Their primary concern in designing the degenerate oligo~ucleotide encoding their variable region was to ensure that all twenty amino acids were encodible at each position; a ~econdary consideration was minimizing the frequency of occurrence of stop ~ignals. Consequently, Scott and Smith and Cwirla et al. employed NNK (N,equal mixture of G, A, T, C; K.equal mixture of G and T) while Devlin et al. used NNS (S~equal mixture of G and C). There was no attempt to minimize the frequency ratio of most favored-to-lea~t favored amino acid, or to equalize the rate of occurrence of acidic and basic amino acids.
Devlin et al. characterized several affinity-selected streptavidin-binding peptides, but did not measure the affinity constants for these peptides.
Cwirla et al. did determine the affinity constant for his peptides, but were disappointed to find that his best hexapeptides had affinities (350-300nM), "orders of magnitude" weaker than that of the native Met-enkephalin epitope (7nM) recognized by the target antibody. Cwirla et al. speculated that phage bearing peptides with higher affinities remained bound under acidic elution, possibly because of multivalent interactions between phage (carry-ing about 4 copies of pIII) and the divalent target IgG.
Scott and Smith were able to find peptides who9e affinity for the target ~ntibody (A2) was comparable to that of `

~:
`: ` ` ` : ` "
`:

W092/15679 PCT/US92/01;39
2`70~3Q~

the reference myohemerythrin epitope (50nM). However, Scott and Smith likewise expressed concern that some high-affinity peptides were lost, possibly through irreversible binding of fusion phage to target.
Ladner, et al, W090/02809, incorporated by reference herein, describe a process for the generation and identification of novel binding proteins having affinity for a predetermined target. In this process, a gene encoding a potential binding domain (as distinct from a mere epitopic peptide), said gene being obtained by random mutagenesis of a limited number of predete~mined codons, is fused to a genetic element which causes the resulting chimeric expression product to be displayed on the outer surface of a virus (especially a filamentous phage) or a cell. Chromatographic selection i9 then used to identify viruses or cells whose genome includes such a fused gene which coded for the protein which bound to the chromatographic target. Ladner, et al. discusss several methods of recovering the gene of interest when the viruse~ or cells is 90 tightly bound to the target that it cannot be washed off in viable form. These are growing them in situ on the chromatographic matrix, fragmenting the matrix and using it as an inoculant into a culture ve~sel, degrading the linkage between the matrix and the ~arget material, and degrading the viruses or cells but then recovering their DNA. However, these methods will al~o recover viruses or cells which are nonspecifically bound to the target material.
W090/02809 also addressed strategies for mutagenesis, including one which provides all twenty amino acids in substantially equal proportions, but only in the context of mutagenesis of protein domains, not epitopic peptides.

W O 92/156~9 PC~r/US9'/01539 The present invention is intended to overcome the deficiencies discussed above. In one embodiment of the invention, a library of "display phage" is used to identify binding domains with a high affinity for a predetermined target. Potential binding domains are displayed on the surface of the phage. This i8 achieved by expressing a fused gene which encodes a chimeric outer surface protein comprising the potential binding domain and at least a functional portion of a coat protein native to the phage. The preferred embodiment uses a pattern of semirandom mutagenesis, called "variegation", that focuses mutations into those residues of a parental binding domain that are most likely to affect its binding properties and are least likely to destroy its underlying structure. As a result, while any one phage displays only a single foreign binding domain (though possibly in multiple copies), the phage library collectively displays thousands, even millions, of different binding domains.
The phage library is screened by affinity ~eparation techniques to identify those phage bearing successful (high affinity) binding domains, and these phage are recovered and characterized to determine the sequence of the successful binding domains. The3e successful binding domains may then serve as the parental binding domains for another round of variegation and affinity separation.
In another embodiment of the invention, the display phage di~play on their surface a chimeric outer surface protein comprising a functional portion of a native outer surface protein and a potential epitope. In an epitope library made of these display phage, the region corresponding to the foreign epitope is hypervariable.
The library iq screened with an antibody or other binding protein of interest and high affinity epitopes are identified. References to di9play, mutagenesis and ~creening of potential binding domains should be taken to .

W092/1~679 PCT/~S92/01~39 2 ~ O ~ ~ ~3 .~

apply, mutatis mutandis, to display, mutagenesis and ~creening of potential epitopes, unless stated otherwise.
AB previously mentioned, when several copies of the chimeric coat protein are displayed on a single phage, there is a risk that irreversible binding will occur, especially if the target is multivalent (as with an antibody). In thi~ case, the phage last eluted by an elution gradient will not be the ones bearing the highest affinity epitopes or bindi~g domains, but rather will be those having an affinity high enough to hold on to the target under the initial elution condition~ but not 90 `:
high as to bind irreversibly. As a result, the methodology known in the art may fail to recover very high affinity epitopes or binding domains, which for many purposes are the most desirable species.
We propose to cope with the problem of irreversible binding by incorporating into the chimeric coat protein a linker sequence, between the foreign epitope or binding domain, and the sequence native to the wild-type phage coat protein, which is cleavable by a site-specific protease. In this case, the phage library is incubated with the immobilized target. Lower affinity phage are eluted off the target and only the solid phase (bearing the high affinity phage) is retained. The aforementioned linker sequence is cleaved, and the phage particles are released, leaving the bound epitope or binding domain behind. One may then recover the particles (and sequence their DNA to determine the ~equence o~ the corresponding epitope or binding domain) or the bound peptide (and sequence its amino acid3 directly). The former recovery method is preferred, a~ the encoding DNA may be amplified in vitro using PCR or in vivo by transfecting suitable host cells with the high affinity display phage. While the production of fusion protein~ with c:lea~able linkers is known in the art, the use of such linkers to . . ..
','' .. . .

W092/l~679 PCT/US92/01~39 facilitate controlled cleavage of a chimeric coat protein of a phage has not previously been reported.
Another method of addressing irre~ersible binding is appropriate when the binding domains are "mini-protein9", i.e., relati~ely small peptides whose stability of structure primarily attributable to the presence of one or more covalent crosslinks, e.g., disulfide bonds. As in the example above, low affinity phage are remo~ed first. The remaining, high affinity, bound phage are then treated with a reagent which breaks the crosslink, such as dithiothreitol in the case of a domain with disulfide bonds, but does not cleave peptidyl bonds or modify the side chains of amino acids which are not crosslinked. This will usually result in sufficient denaturation to either release the phage outright or to permit their elution by other means.
These two methods, of course, are not mutually exclusive.
In the previously known epitope display phage libraries, the phage genome was altered by rep~acing the gene encoding the wild-type gene III protein of M13 with one encoding a chimeric coat protein. As a result, the five nonmal copies of the wild type gene III protein were all replaced by the chimeric coat protein, whereby each phage had five potential binding sites for the target, and hence a very high potential avidity. With high affinity epitopes (or binding domains), this might well contribute to irreversible binding.
One method of the present invention of reducing the avidity of display phage, especially epitope display phage, for their target, and hence of alle~iating the problem of irreversible binding, is to engineer the phage to contain two genes that each express a coat protein, one e~coding the wild type coat protein, and the other the cognate chimeric coat protein. Thus, phage bearing identical epitopes or binding domains may yet bear . .
, . ~ .

W092/1~679 PCT/US92/01~39 different ratios of wild type to chimeric coat protein molecules, and hence have different avidities. The average ratio for the library will be dependent on the relative levels of expression of the two cognate genes.
It may be advantageou~ to be able to modulate the ratio of the chimeric coat protein to its cognate wild-type coat protein. For example, early in the evolutionary process, the affinity of the binding domains for their target may be rather low, especially if they are based on a parental binding domain which has no affinity for the target.
Modulation may be achieved by placing the chimeric gene under the control of a reaulatable promoter.
While it may be possible to place the cognate wild-type gene under the control of a second, differentlyregulated promoter, this may be impracticible if, as with the Ml3 geneIII, the gene is part of a polycistronic operon. In this case, expression of the wild-type gene may be reduced by replacing its methionine initiation codon with a leucine initiation codon.
Ml3 gene III, as previously noted, encodes one of the minor coat proteins of this filamentous phage (five copies per phage). In view of the difficulties with irreversible binding reported by those modifying this gene 80 that a foreign epitope i8 displayed on the phage coat, use of the Ml3 malor coat protein was clearly discouraged. However, we have found that chimeric major coat proteins are in fact useful for displaying potential binding domains for screening purposes even though (indeed, sometimes because) there are over a thousand copie~ of this protein per phage. It is believed that the major (VIII) coat protein would likewise be useful in constructing an epitope phage library.
We have also developed a linker suitable for attaching potential binding domains (or epitopic .
.. . .
3~ ~ .
~- 10 peptides) to this major coat protein, and perhap~ to other proteins as well.
Finally, to the extent that ~ome of the problems experienced with epitope libraries have been attributable to ~he use of patterns of mutagenesis which lead to highly biased allocations of amino acids, the present invention is also directed to a variety of improved patterns that lead to less biased and hence more efficient epitope phage libraries.

BRIEF DESCRIP~ION OF T~l~: DRAWI~GS
Fi~ure 1 shows how a phage may be used a~ a genetic `
phage. At (a) we have a wild-type precoat protein lodged in the lipid bilayer. The signal peptide is in the periplasmic space. At (b), a chimeric precoat protein, with a potential binding domain interposed between the ~ignal peptide and the mature coat protein sequence, is similarly trapped. At (c~ and (d), the signal peptide has been clea~ed off the wild-type and chimeric proteins, respectively, but certain residues of the coat protein sequence interact with the lipid bilayer to prevent the mature protein from passing entirely into the periplasm.
At (e) and (f), mature wild-type and chimeric protein are assembled into the coat of a single stranded DNA phage as it emerges into the periplasmic space. The phage will pass through the outer membrane into the medium where it can be recovered and chromatographically evaluated.
Fiqure 2 shows the C~s of the coat protein of phage fl.

DETAI~ED DESCRIPTION OF T~E PREF~RRED EM3ODIMENTS
I. DISP~AY STRATEGY
A. General Considerations The present invention contemplates that a potential binding domain (pbd) or a potential epitope will be dis-played on the surface of a phage in the form of a fusion with a coat (outer surface) protein (OSP) of the ph~ge.

- . . . ~ . .

.
.. .. . .~ .. . : :

WO92/1~679 PCT/US92/01539 2 ~ J

This chimeric outer surface protein i8 the processed product of the polypeptide expressed by an di~play gene inserted into the phage genome; therefore: 1) the genome of the phage must allow introduction of the display gene either by tolerating additional genetic material or by having replaceable genetic material; 2) the virion must be capable of packaging the genome after accepting the insertion or substitution of genetic material, and 3) the display of the OSP-IPBD protein on the phage surface must not disrupt virion structure sufficiently to interfere with phage propagation.
When the viral particle is assembled, its coat proteins may attach themselves to the phage: a) from the cytoplasm, b) from the periplasm; or c) from within the lipid bilayer. The immediate expres3ion product of the display gene must feature, at its amino terminal, a functional secretion signal peptide, such as the phoA
signal (MKQSTIALA~LP~FTPVTKA), if the coat protein attaches to the phage from the periplasm or from within the lipid bilayer. If a secretion signal is necessary for the display of the potential binding domain, in an especially preferred embodiment the bacterial cell in which the hybrid gene is expressed is of a "secretion-permissive" strain.
The DNA sequence encoding the foreign epitope or binding domain should precede the sequence encoding the coat protein proper if the amino terminal of the processed coat protein is normally its free end, and should follow it if the carboxy terminal is the normal free end.
The morphogenetic pathway of the phage determines ~he environment in which the IP~D will have opportunity to fold. Periplasmically as~embled phage are preferred when IPBDs contain es~ential disulfides, as such IP~Ds may not fold within a cell (these proteins may fold after the phage is released from the cell). Intracellularly .~ . . . ' ~ .

W092~2~679 PCT/VS92/01~39 .~

assembled phage are preferred when the IPBD needs large or insoluble prosthetic groups (such as Fe4S4 clusters), since the IPBD may not fold if secreted because the prosthetic group is lacking.
When variegation is introduced, multiple infections could generate hybrid GPs that carry the gene for one PBD
but have at lea~t some copies of a different PBD on their surfaces; it is preferable to minimize this possibility by infecting cells with phage under conditions resulting in a low multiple-of-infection (MOI).
For a given bacteriophage, the preferred OSP is usually one that is present on the phage surface in the largest number of copies, as this allows the greatest flex~bility in varying the ratio of OSP-IPBD to wild type OSP and also gives the highest likelihood of obtaining satisfactory affinity separation. Moreover, a protein present in only one or a few copies usually performs an essential function in morphogenesis or infection;
mutating such a protein by addition or insertion is likely to result in reduction in viability of the GP.
Nevertheless, an OSP such as M13 gIII protein may be an excellent choice as OSP to cause display of the PBD.
It is preferred that the wild-type osp gene be pre-served. The i~k~ gene fragment may be inserted either into a ~econd copy of the recipient 09p gene or into a novel engineered 09p gene. It is preferred that the osp-ipbd gene be placed under control of a regulated promoter. Our process for~es the evolution of the PBDs derived from IPBD 90 that some of them develop a novel function, viz. binding to a chosen target. Placing the gene that i5 subject to evolution on a duplicate gene is an imitation of the widely-accepted 9cenario for the evolution of protein families. It i9 now generally accepted that gene duplication is the fir9t step in the evolution of a protein family from an ancestral protein.
By having two copies of a gene, the affected - . ,.
, .. . : .: . . :

--- - . . . :,: . . : , .: . ~. . ~ : . . . : - . ' .~ ,:
: . ~ . : .
. . . ~ . : ..

, WO92/15679 PCT/US92/0~539 21~w~-3 physiological process can tolerate mutations in one of the genes. This process is well understood and documented for the globin family (cf. DICK83, p65ff, and CREI84, pll7-125) .
The user must choose a site in the candidate OSP
gene for inserting a i~bd gene fragment. The coats o~
most bacteriophage are highly ordered. Filamentous phage can be described by a helical lattice; isometric phage, by an icoqahedral lattice. Each monomer of each major coat protein sits on a lattice point and makes defined interactions with each of its neighbors. Proteins that fit into the lattice by making some, but not all, of the normal lattice contacts are likely to destabilize the virion by: a) aborting formation of the virion, b) making the virion unstable, or c) leaving gaps in the virion so that the nucleic acid i9 not protected. Thus in bacteriophage, it is important to retain in engineered OSP-IPBD fusion proteins those residues of the parental OSP that interact with other proteins in the virion. For M13 gVIII, we prefer to retain the entire mature protein, while for M13 gIII, it might suffice to retain the last 100 residues (or even fewer). Such a truncated gIII
protein would be expressed in parallel with the complete gIII protein, as gIII protein is required for phage infectivity.
The display gene i9 placed downstream of a known promoter, preferably a regulated promoter such as lac~V5, tac, or trp.
B. Filamentou~ Phages The filamentous phages, which include M13, fl, fd, Ifl, Ike, Xf, Pfl, and Pf3, are of particular int~rest.
The entire life cycle of the filamentous phage M13, a common cloning and sequencing vector, is well understood.
The genetic structure tSCHA78) of M13 is well known as is the physical structure of the virion (BANN81, BOEK80, CHAN79, ITOK79, KAPB78, KUHN85b, RUHN87, MAKO80, MARV78, -:
.
: . . ' .
.

W092/1~679 PCT/US9~/01539 1~

MESS78, OHKA81, RASC86, RUSS81, SCHA78, SMIT85, WEBS78, and ZIMM82); see RASC86 for a recent review of the structure an~ function of the coat proteins.
Marvin and collaborators (MARV78, MAK080, BANN81) have determined an approximate 3D virion structure of the closely related phage fl by a combination o~ genetics, biochemistry, and X-ray diffraction from fibers of the virus. Figure 2 is drawn after the model of Banner et al. (BANN81) and shows only the Cas of the protein. The apparent holes in the cylindrical sheath are actually filled by protein side groups so that the DNA within is protected. The amino terminus of each protein monomer is to the outside of the cylinder, while the carboxy terminus is at smaller radius, near the DNA. Although other filamentous phages (e.q. Pfl or Ike) have different helical ~ymmetry, all have coats composed of many short ~-helical monomers with the amino terminus of each monomer on the virion surface.
1. M13 Major Coat Protein (gVIII) The major coat protein of M13 is encoded by gene VIII. The 50 amino acid mature gene VIII coat protein is synthesized as a 73 amino acid precoat (SCHA78; ITOK79).
The first 23 amino acids oonstitute a typical signal-sequence which causes the nascent polypeptide to be inserted into the inner cell membrane. Whether the precoat inserts into the membrane by itself or through the action of host ~ecretion components, such as SecA and SecY, remains controversial, but has no effect on the operation of the present invention.
An E. coli signal peptidase (SP-I) recognizes amino acids 18, 21, and 23, and, to a lesser extent, residue 22, and cuts between residues 23 and 24 of the precoat (KUHN85a, KUHN~5b, OLIV87). After removal of the signal 3equence, the amino terminus of the mature coat is located on the periplasmic 9ide of the inner membrane;
the carboxy terminus is on the cytoplasmic side. About .
.. . . .
.. : : , ' ~ .: :: . , '. . . ', , ' . .

W092/1~679 ~_~3 3~ PCT/~'S92/01~39 3000 copies of the mature 50 amino acid coat protein associate side-by-~ide in the inner mem~rane.
We have constructed a tripartite gene comprising:
1) DNA encoding a signal sequence directing secretion of parts (2) and t3) through the inner membrane, 2) DNA encoding the mature BPTI sequence, and 3) DNA encoding the mature Ml3 gVIII protein.
This gene causes BPTI to appear in active form on the surface of Ml3 phage.
2. Ml3 Minor Coat Proteins, Generally An introduced binding domain or epitope may also be displayed on a filamentous phage as a portion of a chimeric minor coat protein. These are encoded by genes III, VI, VII, and IX, and each is present in about 5 copieq per virion and i9 related to morphogene~i~ or infection. In contrast, the major coat protein is present in mor~ than 2500 copies per virion. The gene III, VI, VII, and IX proteins are present at the ends of the virion.
3. The Ml3 gIII Minor Coat Protein The single-stranded circular phage DNA associates with about five copies of the gene III protein and is then extruded through the patch of membrane-a~sociated coat protein in such a way that the DNA is encased in a helical sheath of protein (WEBS78). The DNA does not base pair (that would impose severe restriction~ on the virus genome); rather the bases intercalate with each other independent of sequence.
Smith (SMIT85) and de la Cruz et al. (DELA88) have shown that insertionq into gene II cau~e novel protein domains to appear on the virion outer surface. The mini-protein' R gene may be fused to gene III at the site used by Smith and by de la Cruz et al., at a codon correspond-ing ~o another domain boundary or to a surface loop of the protein, or to the amino terminus of the mature protein.

.

WO92/15679 PcT/us92/ol~39 `.3~`

All published works use a vector containing a single rnodified gene III of fd. Thus, all five copies of gIII
are identically modified. Gene III is quite large (1272 b.p. or about 20~ of the phage genome) and it is uncertain whether a duplicate of the whole gene can be stably inserted into the phage. Furthermore, all five copies of gIII protein are at one end of the virion.
When bivalent target molecules (such as antibodies) bind a pentavalent phage, the resulting complex may be irreversible. Irreversible binding of the phaye to the target greatly interfereq with affinity enrichment of the GPs that carry the genetic sequences encoding the novel polypeptide having the highest affinity for the target.
To reduce the likelihood of formation of irreversible complexes, we may use a second, synthetic gene that encodes only the carboxy-terminal portion of ~II- We might, for example, engineer a gene that comprises ~from 5' to 3'):
1) a promoter (preferably regulated), 2) a ribosome-binding site, 3) an initiation codon,
4) a sequence encoding a functional signal peptide directing secretion of parts (5) and (6) through the inner membrane,
5) DNA encoding a potential binding domain,
6) DNA encoding residues 275 through 424 of M13 gIII
protein,
7) a translation stop codon, and
8) (optionally) a transcription stop signal.
Note that in the gIII protein, the amino terminal moiety is re~ponsible for pilus binding (i.e., for infecti~ity) and the carboxy terminal moiety for packaging, so that the chimeric gIII protein described above is able to asRemble into the viral coat, but does not contribute to infectivity.

, .

. .

WO92/lS679 PCT~US92/01~9 2~ Q~3~?~

We leave the wild-type gene III 80 that some unaltered gene III protein will be present.
Thus, the hybrid gene may comprise DNA encoding a potential binding domain operably linked to a ~ignal sequence (e.g., the signal sequences of the bacterial phoA or bla genes or the signal ~equence of M13 phage qene~) and to DNA encoding at least a functional portion of a coat protein (e.q., the M13 gene III or gene VIII proteins) of a filamentous phage (e.q., M13). The expression product is transported to the inner membrane (lipid bilayer) of the host cell, whereupon the signal peptide is cleaved off to leave a processed hybrid protein. The C-terminus of the coat protein-like com-ponent of this hybrid protein is trapped in the lipid bilayer, so that the hybrid protein does not escape into the periplasmic space. (This i9 typical of the wild-type ~oat protein.) As the single-stranded DNA of the nascent phage particle passes into the periplasmic space, it collects both wild-type coat protein and the hybrid protein from the lipid bilayer. The hybrid protein is thus phaged into the surface sheath of the filamentous phage, leaving the potential binding domain expo~ed on its outer surface.
4. Coat Proteins of Pf3 Similar constructions could be made with other filamentous phage. Pf3 is a well known filàmentous phage that infects Pseudomonas aeruqenosa cells that harbor an IncP-1 plasmid. The entire genome has been sequenced (~UIT85) and the genetic signals involved in replication and as~embly are known (LUIT87). The major coat protein of PF3 is unusual in having no signal peptide to direct its secretion. The sequence ha~ charged residues ASP~, ARG37, LYS40, and PHE44-COO-which is consi~tent with the amino terminus being exposed. Thus, to cause an IPBD to appear on the surface of Pf3, we construct a tripartite gene comprising:

W092/1~679 PCT/US92/01539 ' l) a signal sequence known to cause secretion in P.
aer~qenosa (preferably known to cause secretion of IPBD) fused in-frame to, 2) a gene fragment encoding the IPBD ~equence, fused in-frame to, 3) DNA encoding the mature Pf3 coat protein.
Optionally, DNA encoding a flexible linker of one to lO
amino acids is introduced between the ipbd gene fragment and the Pf3 coat-protein gene. Optionally, DNA encoding the recognition site for a specific protease, such as tissue plasminogen activator or blood clotting Factor Xa, is introduced between the ipbd gene ~ragment and the Pf3 coat-protein gene. Amino acids that form the recognition site for a specific protease may also serve the function of a flexible linker. This tripartite gene is introduced into Pf3 so that it does not interfere with expression of any Pf3 genes. To reduce the possibility of genetic recombination, part (3) is designed to have numerous silent mutations relative to the wild-type gene. Once the signal sequence is cleaved off, the IPBD is in the periplasm and the mature coat protein acts as an anchor and phage-assembly signal. It does not matter that this fusion protein comes to rest in the lipid bilayer by a route different from the route followed by the wild-type coat protein.
Gene Co~a~ction The structural coding sequence of the display gene encodes a chimeric coat protein and any required ~ecretion signal. A "chimeric coat protein" is a fusion of a first amino acid sequence (essentially corresponding to at least a functional portion of a phage coat protein) with a second amino acid sequence, e.g., a domain foreign to and not substantially homologous with any domain of the first protein. A chimeric protein may present a foreign domain which is found (albeit in a different pro~ein) in an organism which also expresses the first - ~: ' ' .

W092~15679 PcT/us9~/ol~39 ~ 1 ~3 ~

protein, or it may be an "interspecies", "intergenericn, etc. fusion of protein structures expre~sed by different kinds of organisms. The foreign domain may appear at the amino or carboxy terminal of the first amino acid sequence (with or without an intervening spacer), or it may interrupt the first amino acid sequence. The first amino acid sequence may correspond exactly to a surface protein of the phage, or it may be modified, e.g., to facilitate the display of the binding domain.
A preferred site for insertion of the i~bd gene into the phage ocp gene is one in which: a) the IP9D folds into its original shape, b) the OSP domains fold into their original shapes, and c) there is no interference between the two domains.
If there is a model of the phage that indicates that either the amino or carboxy terminus of an OSP i8 exposed to solvent, then the exposed terminus of that mature OSP
becomes the prime candidate for insertion of the ipbd gene. A low resolution 3D model suffices.
In the absence of a 3D structure, the amino and carboxy termini of the mature OSP are the best candidates for insertion of the ipbd gene. A functional fusion may require additional residues between the IPBD and OSP
domains to avoid unwanted interactions between the domain~. Random-sequence DNA or DNA coding for a specific sequence of a protein homologous to the IPBD or OSP, can be inserted between the o~p fragment and the ipbd fragment if needed.
Fusion at a domain boundary within the OSP i~ also a good approach for obtaining a functional fusion. Smith exploited such a boundary when subcloning heterologous DNA into gene III of fl (SMIT~5).
The criteria for identifying OSP domains suitable for causing display of an IPBD are somewhat different from those used to identify and IPBD. When identifying an OSP, minimal size is not so important because the OSP

W092/15679 PCT/US92/01~39 _~ ~ C`, J

domain will not appear in the final binding molecule nor will we need to synthesize the gene repeatedly in each variegation round. The major design concerns are that:
a) the OSP::IPBD fusion causes display of IPBD, b) the initial genetic construction be reasonab}y convenient, and c) the 08pt: ipbd gene be genetically stable and easily manipulated. There are several methods of identifying domains. Methods that rely on atomic coordinates have been re~iewed by Janin and Chothia ~JANI85). These methods use matrices of distances between ~ carbons (Ca), di~iding planes (cf. ROSE85), or buried surface (RASH84). Chothia and collaborators have correlated the behavior of many natural proteins with domain structure (according to their definition). Rashin correctly predicted the stability of a domain comprising residues 206-316 of thermolysin ~VITA84, RAS~84).
Many researchers ha~e used partial proteolysis and protein sequence analysis to isolate and identify stable domains. (See, for example, VITA84, POTE83, SCOT87a, and PA~079.) Pabo ~ al. used calorimetry as an indicator that the cI repressor from the coliphage )\ contains two domains; they then used partial proteolysis to determine the location of the domain boundary.
If the only structural information available is the 2s amino acid seguence of the candidate OSP, we can u~e the sequence to predict turns and loops. There is a high probability that some of the loops and turns will be correctly predicted (cf. Chou and Fasman, ~CHOU74));
these locations are also candidates for insertion of the ipb~ gene fragment.
Fusing one or more ~ew domains to a protein may make the ability of the new protein to be exported from the cell different from the ability of the parental protein.
The signal peptide of the wild-type coat protein may function for authentic polypeptide but be unable to direct export of a fusion. To utilize the Sec-dependent '" ' ' '~ ~ ' ~ . .

W092tl5679 PCTIUS92/01~39 2 ~ J

pathway, one may need a different signal peptide. Thus, to express and display a chimeric BPTI/M13 gene VIII
protein, we found it necessary to utilize a heterologous signal peptide (that of ~hoA).
s Phage that display peptides having high affinity for the target may be quite difficult to elute from the target, particularly a multivalent target. One can introduce a cleavage site for a specific protease, such as blood-clotting Factor Xa, into the fusion protein 90 that the binding domain can be cleaved from the genetic package. Such cleavage has the advantage that all resulting phage have identical coat proteins and therefore are equally infective, even if polypeptide-displaying phage can be eluted from the affinity matrix without cleavage. This step allows recovery of valuable genes which might otherwiae be lost. To our knowledge, no one has disclosed or suggested using a specific protease as a means to recover an information-containing genetic package or of converting a population of phage that vary in infectivity into phage having identical infectivity.
There exist a number of highly specific proteases.
While the in~ention does not reside in the choice of any particular protease, the protease is preferably sufficiently specific 80 that under the cleavage conditions, it will cleave the linker but not any polypeptide essential to the viability of the phage, or (save in rare cases) the potential epitope/binding domain. It is possible that choice of particular cleavage conditions, e.g., low temperature, may make it feasible to use a protease that would otherwise be unsuitable.
The blood-clotting and complementation systems contains a number of very specific proteases. Usually, 3~ the enzymes at the early stages of a cascade are more specific than are the later ones. For example, Factor X, W092/1~679 PCT/US92/01539 (F.X,) is more specific than is thrombin (cp. Table 10-2 of C0LM87). Bovine F.X, cleaves after the sequence Ile-Glu-Gly-Arg while human F.X, cleaves after Ile-A~p-Gly-Arg. Either protea~e-linker pair may be used, as desired. If thrombin is used, the mQst preferred thrombin-sensitive linkers are those found in fibrinogen, Factor XIII, and prothrombin. Preferably, one would take the linker sequence from the species from which the thrombin is obtained; for example, if bovine thrombin is to be used, then one uses a linker taken from bovine fibrinogen or bovine F.XIII.
Human Factor XI, cleaves human Factor IX at two places (C0LM87, p.42):
Q T S K ~ T R~4~A E A V F and S F N D F T R~/ V V G G E.
Thus one could incorporate either of these sequences te9pecially the underscored portions) as a linker between the PBD and the GP-surface-anchor domain (GPSAD) and use human F.XI, to cleave them.
Human kallikrein cuts human F.XII at R353 (C0LM87, p.258):
L F S S M T R353/ V V G G ~ V.
This sequence has significant similarity to the hF.XI, sites above. One could incorporate the ~equence SSMTRW G
as a linker between PBD and GPSAD and cleave PBD from the GP with human kallikrein.
Human F.XII, cuts human F.XI at R369 (C0LM87, p.256):
X I ~ P R~69/ I V G G T.
One could incorporate KI~PRIVG as a linker between P~D
and GPSAD. P~D could then be cleaved from GP with hF.XII,.
Other proteases that have been used to clea~e fusion proteins include enterokinase, collagenase, chymosin, urokinase, renin, and certain signal peptidases. See Rutter, US 4,769,326.

', . :, .

Wo92/15679 PCT/~IS92/01~39 When a protease inhibitor i8 sought, the target protea~e and other proteases having similar ~ubstrate ~pecificity are not preferred for cleaving the P~D from the GP. It i9 preferred that a linker resembling the substrate o~ the target protease ~Q be incorporated anywhere on the display phage because this could make separation of excellent binders from the rest of the population needlessly more difficult.
If there is steric hindrance of the site-specific cleavage of the linker, the linker may be modified ~o that the cleavage site is more exposed, e.g., by interposing glycines (for additional flexibility) or prolines (for maximum elongation) between the cleavage site and the bulk of the protein. GUAN91 improved thrombin cleavage of a GST fusion protein by introducing a glycine-rich linker (PGISGGGGG) immediately after the thrombin cleavage site (~VPRGS). A suitable linker may also be identified by variegation-and-selection.
The sequences of regulatory parts of the gene are ~0 taken from the sequences of natural regulatory elements:
a) promoters, b) Shine-Dalgarno sequences, and c) trans-criptional terminators. Regulatory elements could alsobe designed from knowledge of consensus sequences of natural regulatory regions. The sequence~ of these regulatory element~ are connected to the codiny regions;
reqtriction sites are also inserted in or adjacent to the regulatory regions to allow convenient manipulation.
The essential function of the affinity separation is to separate GPs that bear PBDs (derived from IPBD) having high affinity for the target from GPs bearing P~Ds having low affinity for the target. If the elution volume of a phage depends on the number of PBDs on the phage surface, then a phage bearing many PBDs with low affinity, GPtPBD~), might co-elute with a phage bearing fewer PBDs with high affinity, GP(PBD,). Regulation of the display gene preferably is such that most phage display WO92/1~679 PCT/US92/Ot~39 j3 ~ufficient PBD to effect a good separation according to affinity. Use of a regulatable promoter to control the level of expression of the display gene allow~ fine adjustment of the chromatographic behavior of the ~ariegated population.
Induction of synthe~is of engineered genes in vegetative bacterial cells has been exercised through the use of regulated promoters such as lac W5, trpP, or (MANI82). The factors that regulate the quantity of protein syntheYized are sufficiently well understood that a wide variety of heterologous proteins can now be produced in E. ~Qli, ~ ubtilis and other host cells in at least moderate quantities (SKER88, BETT88).
Preferably, the promoter for the display gene is ~ubject lS to regulation by a small chemical inducer. For example, the lac promoter and the hybrid ~Ep-lac (~ac) promoter are regulat~ble with isopropyl thiogalactoside (IPTG).
The promoter for the constructed gene need not come from a natural osp gene; any regulatable promoter functional in bacteria can be used. A non-leaky promoter is preferred.
The coding portions of genes to be synthesized are designed at the protein level and then encoded in DNA.
While the primary consideration in devising the DNA
sequence is obtaining the desired diverYe population of potential binding domains (or epitopes), consideration i9 al~o given to providing restriction site~ to facilitate further gene manipulation, minimizing the potential for recombination and spontaneous mutation, and achieving efficient translation in the chosen host cells.
The present invention is not limited to any parti-cular method of DNA syntheYis or construction. Conven-tional DNA synthe3izers may be used, with appropriate reagent modifications for production of variegated DNA
(similar to that now used for production of mixed probes).

- ' `
~:
:, W092/15679 PCT/~lS92/01~39 2 ~

The phage are genetically engineered and then tran~fected into host cells, e.g., E. coli. B. ~ubtili~
or P. aeruginosa. suitable for amplification. The present invention i9 not limited to any one method of S transforming cells with DNA or to any particular host cells.
NIT~ . POTENTIA~ BI~IDING DO~ N (IPBD):
By virtue of the present invention, proteins may be obtained which can bind specifically to targets other than the antigen-combining sites of antibodies. For the purposes of the appended claims, a protein P is a bindinq protein if for some target other than the variable domain of an antibody, the dissociation constant KD (P,A) c 10 moles/liter (preferably, c 10 7 moles/liter). The exclusion of "variable domain of an antibody" is intended to make clear that for the purposes herein a protein is not to be considered a "binding protein" merely because it is antigenic. However, an antigen may nonetheless qualify as a binding protein because it specifically binds to a substance other than an antibody, e.~., an enzyme for its substrate, or a hormone for its cellular receptor. Additionally, it should be pointed out that "binding protein" may include a protein which binds specifically to the Fc of an antibody, e.a., staphylococcal protein A.
While the present invention may be used to develop novel antibodies through variegation of codons corresponding to the hypervariable region of an antibody's variable domain, its primary utility resides in the development of binding proteins which are not antibodies or even variable domains of antibodies. Novel antibodies can be obtained by immunological techniques;
novel enzymes, hormones, e~c. cannot.
Most larger proteins fold into distinguishable globule~ called domains. The display ~trategy is first perfected by modifying a genetic phage to di~play a W092/1~679 ~ PCT/US92/01~39 stable, structured domain (the ~ini~ial DQ~ential binding domainll, IPBD) for which an affinity molecule (which may be an antibody) is obtainable. The success of the modifications i8 readily measured by, e.q., determining whether the modified genetic phage binds to the affinity molecule. For the purpose of identifying IPBDs, definitions of "domain~ which emphasize stability --retention of the overall structure in the face of perturbing forces such as elevated temperatures or chaotropic agents -- are favored, though atomic coor-dinates and protein sequence homology are not completely ignored. When a domain of a protein is primarily responsible for the protein~ ability to specifically bind a chosen target, it is referred to herein as a 15 "binding domain" (BD). ~-The IPBD i8 chosen with a view to its tolerance for extensive mutagenesis. Once it is known that the IPBD
can be displayed on a surface of a phage and subjected to affinity selection, the gene encoding the IP~D is sub-jected to a special pattern of multiple mutagenesis, here termed "variegat~Qn", which after appropriate cloning andamplification steps leads to the production of a popula-tion of phage each of which displays a single potential binding domain (a mutant of the IPBD), but which collectively display a multitude of different though structurally related potential binding domains (PBDs).
Each genetic phage carries the version of the ~ gene that encodes the PBD displayed on the surface of that particular phage. Affinity selection is then used to identify the display phage bearing the PBDs with the desired binding characteristics, and the9e di9play phage may then be amplified. After one or more cycles of enrichment by affinity selection and amplification, the DNA encoding the successful binding domains (SBDs) may then be recovered ~rom selected phage.

' ' . .
- ' .
. . , . -..
.

wo 92/1~679 ~r/VS9'/OtS39 ,? ~`i 'tf' If need be, the DNA from the SBD-bearing phage may then be further "~ariegated", using an SBD of the la~t round of variegation as the ~parental potential binding domain~ (PP~D) to the next generation of PBDs, and the process continued until the worker in the art is satisfied with the result. At that point, the SBD may be produced by any conventional means, including chemical synthesis.
The initial potential binding domain may be: 1) a domain of a naturally occurring protein, 2) a non-natur-ally occurring domain which substantially corresponds in sequence to a naturally occurring domain, but which differ~ from it in sequence by one or more substitutions, insertions or deletions, 3) a domain substantially corresponding in sequence to a hybrid of subsequences of two or more naturally occurring proteins, or 4) an artificial domain designed entirely on theoretical grounds based on knowledge of amino acid geometries and statistical evidence of secondary structure preferences of amino acids. (However, the limitations of a ~riori protein design prompted the present invention.) Usually, the domain will be a known binding domain, or at least a homologue thereof, but it may be derived from a protein which, while not possessing a known binding activity, possesses a secondary or higher structure that lends itself to binding activity (clefts, grooves, etc~). The protein to which the IPBD is related need not have any ~pecific affinity for the target material.
In determining whether sequences should be deemed to "substantially correspond", one should consider the following issues: the degree of sequence ~imilarity when the ~equences are aligned for best fit according to standard algorithms, the similarity in the connectivity patterns of any crosslinks (e.q., disulfide bonds), the degree to which the proteins have similar three-dimen-sional structures, as indicated by, e.g., X-ray diffrac-WO92/1~679 PCT/US92/01539 , ~, ~,, .
~ ~;~`'` " `

tion analysis or NMR, and the degree to which the se-quenced proteins have similar biological activity. In this context, it should be noted that among the serine protease inhibitors, there are families of proteins recognized to be homologous in which there are pairs of members with as little as 30~ sequence homology.
A candidate IPBD should meet the following criteria:
1) a domain exists that will remain stable under the conditions of its intended use (the domain may comprise the entire protein that will be inserted, e.g. BPTI, ~-conotoxin GI, or CMTI-III), 2) knowledge of the amino acid sequence is obtain- -`
able, and 3) a molecule is obtainable having specific and high affinity for the IPBD, AfM(IPBD).
Preferably, in order to guide the variegation strategy, knowledge of the identity of the residues on the domain's outer surface, and their spatial relationship~, is obtainable; however, this consideration is less important if the binding domain is small, e.q., under 40 residues.
Preferably, the IPBD is no larger than necessary because small SBDs (for example, less than 40 amino acids) can be chemically synthesized and because it is easier to arrange restriction sites in smaller amino-acid sequences. For PBDs smaller than about 40 residues, an added advantage is that the entire variegated E~ gene can be synthesized in one piece. In that case, we need arrange only suitable restriction sites in the osp gene.
A smaller protein minimize~ the metabolic strain on the 3~ phage or the host of the GP. The IPBD is preferably smaller than about 200 residues. The IPBD must also be large enough to have acceptable binding affinity and specificity. For an IPBD lacking covalent crosslinks, such as disulfide bonds, the IPBD is preferably at least 40 residues; it may be as small as 5iX residues if it contains a crosslink.

.
.
' " ' ~ '' :
: .
.

WO92/1~679 PCT/US92/01539 2 ~

There are many candidate IPBDs, for example, bovine pancreatic trypsin inhibitor (BPTI, 58 residues), CMTI-III t29 residues), crambin (46 residues), third domain of ovomucoid (56 residues), heat-stable enterotoxin (ST-Ia of E. coli) (13 residues), ~-Conotoxin GI (13 residues), ~-Conotoxin GIII (22 residues), Conus King ~ong mini-protein (27 residues), T4 lysozyme (164 residues), and azurin (128 residues). Table 50 lists several preferred IPBDs.
10In some cases, a protein having some affinity for the target may be a preferred IPBD even though some other criteria are not optimally met. For example, the Vl domain of CD4 is a good choice as IPBD for a protein that binds to gpl20 of HIV. It i~ known that mutations in the 15region 42 to 55 of Vl greatly affect gpl20 binding and that other mutations either have much less effect or completely disrupt the structure of Vl. Similarly, tumor necrosis factor (TNF) would be a good initial choice if one wants a TNF-like molecule having higher affinity for the TNF receptor.
As even surface mutations may reduce the stability of the PBD, the chosen IPBD should have a high melting temperature (50C acceptable, the higher the better; BPTI
melts at 95C.) and be stable over a wide pH range (8.0 25to 3.0 acceptable; 11.0 to 2.0 preferred), so that the SBDs derived from the chosen IPBD by mutation and selection-through-binding will retain ~ufficient stabil-ity. Preferably, the substitutions in the IPBD yielding the various PBDs do not reduce the melting point of the domain below -40 C. Mutations may arise that increase the stability of 5BDs relative to the IPBD, but the process of the present invention does not depend upon this occurring. Proteins containing covalent cros~links, such as multiple disulfides, are usually sufficiently stable.
A protein having at least two disulfides and having at W092/1~679 PcT/us9~ols39 ;Q`~3 3 ~ .

least 1 disulfide for every twenty residues may be presumed to be sufficiently stable.
If the target is a protein or other macromolecule a preferred embodiment of the IPBD i8 a small protein such as the Cucurbita maxima trypsin inhibitor III (29 resi-dues), BPTI from Bos Taurus (58 residues), crambin from rape seed (46 residues), or the third domain of ovomucoid from Coturnix cotuX~ix Japoniça (Japanese quail) (56 residues), because targets from this class have clefts and grooves that can accommodate small proteins in highly specific ways. If the target is a macromolecule lacking a compact structure, such as starch, it should be treated as if it were a small molecule. Extended macromolecules with defined 3D structure, such as collagen, should be treated as large molecules.
If the target i9 a small molecule, such as a steroid, a preferred embodiment of the IPBD is a protein of about 80-200 residues, such as ribonuclease from Bos taurus (124 residues), ribonuclease from Asperqillus oruzae (104 residues), hen egg white lysozyme from Gallus qallus (129 residues), azurin from Pseudomonas aeruqenosa (128 residues), or T4 lysozyme (164 residues), because such proteins have clefts and grooves into which the small target molecules can fit. The ~rookhaven Protein Data Bank contains 3D structures for all of the proteins listed. Genes encoding proteins as large as T4 lysozyme can be manipulated by standard techniques for the purposes of this invention.
If the target is a mineral, insoluble in water, one considers the nature of the molecular surface of the mineral. Minerals that have smooth surfaces, such as crystalline silicon, are be~t addressed with medium to large proteins, such as ribonuclease, as IPBD in order to have sufficient contact area and specificity. Minerals with rough, grooved ~urfaces, such as zeolites, could be . ., ' ; :: .
.

W09~ 679 PCT/US92/01~39 bound either by small proteins, such as BPTI, or larger proteins, such as T4 lysozyme.
~ PTI is an especially preferred IPBD because it meets or exceeds all the criteria: it is a small, very stable protein with a well known 3D structure.
Small polypeptides have potential advantages over larger polypeptides when used as therapeutic or diagnostic agents, including (but not limited to):
a) better penetration into tissues, b) faster elimination from the circulation (important for imaging agents), c) lower antigenicity, and d) higher activity per mass.
Thus, it would be desirable to be able to employ the combination of variegation and affinity selection to identify small polypeptides which bind a target of choice.
Polypeptides of this size, however, have disadvan-tage~ as binding molecules. According to Olivera et al.
(OLIV9Oa): ~Peptides in this size range normally equi-librate among many conformations (in order to have a fixed conformation, proteins generally have to be much larger)." Specific binding of a peptide to a target molecule requires the peptide to take up one conformation that is complementary to the binding site.
In one embodiment, the present invention overcomes the~e problems, while retaining the advantages of smaller polypeptides, by fostering the biosynthesis of novel mini-proteins having the desired binding characteristics.
Mini-Proteins are small polypeptides (usually less than about 60 residue~, more preferably less than 40 residues (~micro-proteinsn)) which, while too small to have a stable conformation as a result of noncovalent forces alone, are covalently crosslinked (e.~., by disulfide bonds) into a stable conformation and hence have biological activities more typical of larger protein .
: ~ -, '- , ~ ..

W092/15679 ~ PCT/US97/Ot~39 molecules than of unconstrained polypeptides of ~omparable size.
When mini-proteins are variegated, the residues which are covalently crosslinked in the parental molecule are left unchanged, thereby stabilizing the conformation.
For example, in the variegation of a disulfide bonded mini-protein, certain cysteines are invariant 90 that under the conditions of expression and display, covalent crosslinks (e.g., disulfide bonds between one or more pairs of cysteines) form, and substantially constrain the conformation which may be adopted by the hypervariable linearly intermediate amino acids. In other words, a constraining scaffolding is engineered into polypeptides which are otherwise extensively randomized.
Once a mini-protein of desired binding character-istics is characterized, it may be produced, not only by recombinant DNA techniques, but also by nonbiological synthetic methods.
For the purpose of the appended claims, a mini-protein has between about eight and about 60 residues.An intrachain disulfide bridge connecting amino acids 3 and 8 of a 16 residue polypeptide will be said herein to have a span of 4. If amino acids 4 and 12 are also disulfide bonded, then their bridge has a span of 7.
Together, the four cysteines divide the polypeptide into four intercysteine segments (1-2, 5-7, 9-11, and 13-16).
tNote that there i9 no segment between Cys3 and Cys4.) The connectivity pattern of a crosslinked mini-protein i~ a simple description of the relati~e location of the termini of the crosslinks. For example, for a mini-protein with two disulfide bonds, the connecti~ity pattern "1-3, 2-4" means that the first crosslinked cysteine is disulfide bonded to the third crosslinked cysteine (in the primary sequence),~and the second to the fourth.

.
.

, W092/1~679 PCT/US9~/Ot~39 ~ 1 Q c~3 3 ~; 3 The variegated disulfide-bonded mini-proteins of the present invention fall into several classes.
Class_I minl-proteins are those featuring a single pair of cysteines capable of interacting to form a disulfide bond, said bond having a span of no more than nine residues. This disulfide bridge preferably has a span of at least two residues; this is a function of the geometry of the disulfide bond. When the spacing is two or three residues, one residue is preferably glycine in order to reduce the strain on the bridged residues. The upper limit on spacing is less precise, however, in general, the greater the spacing, the less the constraint on conformation imposed on the linearly intermediate amino acid residues by the disulfide bond.
A disulfide bridge with a span of 4 or 5 is espe-cially preferred. If the span is increased to 6, the constraining influence i8 reduced. In this case, we prefer that at least one of the enclosed residues be an amino acid that imposes restrictions on the main-chain geometry. Proline imposes the most restriction. Valine and isoleucine restrict the main chain to a lesser extent. The preferred position for this constraining non-cy~teine residue i9 adjacent to one of the invariant cysteines, however, it may be one of the other bridged residues. If the span is seven, we pre~er to include two amino acids that limit main-chain conformation. These amino acids could be at any of the seven positions, but are preferably the two bridged residues that are immediately adjacent to the cysteines. If the span is eight or nine, additional constraining amino acids may be provided.
Additional amino acids may appear on the amino side of the first cysteine or the carboxy side of the second cysteine. Only the immediately proximate "unspanned"
amino acids are likely to have a significant effect on the con~ormation of the span.

.. . .. ..
: . .
. : . : .

W092/1~679 PCT/~S92/01539 1ass II mini-proteins are those featuring a single disulfide bond having a span of greater than nine amino acids. The bridged amino acids form secondary structures which help to stabilize their conformation. Preferably, these intermediate amino acids form hairpin supersecondary structures such as those Rchematized below:
--- S--S
-Cys-~helix-turn-~strand-Cys-o r s--s -Cys-~helix-turn-~helix-Cys--- S--S
-Cys-~strand-turn-~strand-Cys-In designing a suitable hairpin structure, one may copy an actual structure from a protein whose three-dimensional conformation is known, design the structure using secondary structure tendency data for the individual amino acids, etc., or combine the two approaches. Preferably, one or more actual structures are used as a model, and the frequency data is used to determine which mutations can be made without disrupting the structure.
Preferably, no .more than three amino acids lie between the cysteine and the beginning or end of the helix or ~ strand.
More complex structures (such as a double hairpin) are also possible.
Class III mini-proteins are those featuring a plurality of disulfide bonds. They optionally may also feature secondary structures such as those discussed above with regard to Class II mini-proteins. Since the number of possible di~ulfide bond topologies increases rapidly with the number of bonds (two bonds, three topologies; three bonds, 15 topologies; four bonds, 105 topologies) the number of disulfide bonds preferably does -, ' . - ' ' ~ ' - : . .
-.
, . . , :

WO92/1~679 PCTtUS9~/01539 2 ~ 0;

not exceed four. Two disulfide bond are preferable to ~hree, and three to four. With two or more disulfide bonds, the disulfide bridge spans preferably do not exceed 30, and the largest intercysteine chain segment preferably doe~ not exceed 20.
Naturally occurring class III mini-proteins, such as heat-~table enterotoxin ST-Ia, frequently have pairs of cysteines that are clustered (-C-C- or -C-X-C-) in the amino-acid sequence. Clustering reduces the number of realizable topologies,~ and may be advantageous.
Metal Finqer Mini-Proteins. The mini-proteins of the present invention are not limited to those crosslinked by disulfide bonds. Another important class of mini-proteins are analogues of finger proteins.
Finger proteins are characterized by finger structures in which a metal ion is coordinated by two Cy9 and two His re~idues, forming a tetrahedral arrangement around it.
The metal ion i9 most often zinc(II), but may be iron, copper, cobalt, etc. The "finger" has the consensus sequence (Phe or Tyr)-(l AA)-Cys-(2-4 AAs)-Cys-(3 AAs)-Phe-(5 AA5) -Leu-(2 AAs)-His-(3 AAs)-His-(5 AAs)(BERG88;
GIBS88). The present invention encompasses mini-proteins with either one or two fingers.
Further diversity may be introduced into a display phage library ofpotential binding domains by treating the phage with (preferably nontoxic) enzymes and/or chemical reagents that can selectively modify certain side groups o~ proteins, and thereby affect the binding properties of the diRplayed PBDs. Using affinity separation methods, we enrich for the modified GPs that bind the predetermined target. Since the active binding domain is not entirely genetically specified, we must repeat the post-morphogenesis modification at each enrichment round.
Thi~ approach is particularly appropriate with mini-protein IPBDs becau3e we en~ision chemical synthesis ofthese SBDs.

.: . , '
9 PCI`/US9~/01~39 ?.~ 3~ 36 EPI~OPIC PEPTIDES
The present invention also relates to the identification of epitopic peptides which bind to a target which is the epitopic binding site of an antibody, lectin, enzyme, or other binding protein. In the case of an antibody, the epitopic peptide will be at least four amino acids and more preferably at least six or eight amino acids. Usually, it will be les~ than 20 amino acids, but there is no fixed upper limit. In general, however, the epitopic peptide will be a "linear"
or "sequential" epitope. Typically, in constructing a library for displaying epitopic peptides, all or mo~t of the amino acid positions of the potential epitope will be varied. However, it is desirable that among those amino acids allowed at a particular position, that there be a relatively equal repre~entation, as further discussed below in the context of mutagenesis of protein domains.

VARIEGATION STRATEGY -- MnTA&ENESIS TO OBTAIN POTENTIAL
BINDING DOMAINS (OR EPITOPES) WIT~ DESIRED DIYERSITY
When the number of different amino acid sequences obtainable by mutation of the domain is large when compared to the number of different domains which are displayable in detectable amounts, the efficiency of the forced evolution is greatly enhanced by careful choice of which residues are to be varied. First, residues of a know~ protein which are likely to affect its binding activity (e.g., surface residues) and not likely to unduly degrade its stability are identified. Then all or some of the codons encoding these residues are varied simultaneously to produce a variegated population of DNA.
The variegated population of DNA is used to express a variety of potential binding domains, whose ability to bind the target of intere~t may then be evaluated.
The method of the present invention is thus further di~tinguished from other methods in the nature of the . . . . ':
. ,,, ' ''. : . .' .
' - . :

WO9~/15679 PCT/US92/01539 2 ~ 3 highly variegated population that is produced and from which novel binding proteins are selected. We force the displayed potential binding domain to sample the nearby "sequence space" of related amino-acid ~equences in an efficient, organized manner. Four goals guide the various variegation plans used herein, preferably: 1) a very large number (e.q. 107) of variants is a~ailable, 2) a very high percentage of the possible variants actually appears in detectable amounts, 3) the frequency of appearance of the desired variants is relatively uniform, and 4) variation occurs only at a limited number of amino-acid residues, most preferably at residues having side groups directed toward a common region on the surface of the potential binding domain.
This i9 to be distinguished from the simple use of indiscriminate mutagenic agents such as radiation and hydroxylamine to modify a gene, where there is no (or very oblique) control over the site of mutation. Many of the mutations will affect residues that are not a part of the binding domain. Moreover, since at a reasonable level of mutagenesis, any modified codon is likely to be characterized by a single base change, only a limited and biased range of possibilities will be explored. Equally remote is the u~e of site-specific mutagenesi~ techniques employing mutagenic oligonucleotides of nonrandomized seguence, since the~e techniques do not lend themselves to the production and testing of a large number of variants. While focused random mutagenesis techniques are known, the importance of controlling the distribution of variation has been largely overlooked.
The term ~Ivariegated DNA" (vgDNA) refers to a mixture of DNA molecules of the same or similar length which, when aligned, vary at some codons so as to encode at each such codon a plurality of different amino acids, but which encode only a single amino acid at other codon po~itions. It i~ further under~tood that in variegated . . . - . .

W O 92tl5679 ~, PC~r/VS9_/01~39 ~ Q~3 '-DNA, the codons which are variable, and the range and frequency of occurrence of the different amino acids --which a given variable codon encodes, are determined in advance by the synthe~izer of the DNA, even though the ~ynthetic method does not allow one to know, a priori, the sequence of any individual DNA molecule in the mixture. The number of designated variable codons in the variegated DNA i9 preferably no more than 20 codons, and more preferably no more than 5-10 codons. The mix of amino acids encoded at each variable codon may differ from codon to codon. A population of display phage into which variegated DNA has been introduced is likewise said to be "variegated".
When DNA encoding a portion of a known domain of a protein is variegated, the original domain i9 called the parent of the potential binding domains (PPBD), and the multitude of mutant domains encoded as a result of the variegation are collectively called the "potential binding domain~" (PBD), as their ability to bind to the predetermined target i9 not then known.
We now consider the manner in which we generate a diverse population of potential binding domains in order to facilitate selection of a PBD-bearing phage which binds with the requisite affinity to the target of choice. The potential binding domains are first designed at the amino acid level. Once we have identified which residues are to be mutagenized, and which mutations to allow at those positions, we may then design the variegated DNA which is to encode the various PBDs so as to assure that there is a reasonable probability that if a PBD has an affinity for the target, it will be detected. Of course, the number of independent transformants obtained and the sensitivity of the affinity separation technology will impo5e limits on the exten~ of ~ariegation possible within any single round of variegation.

.
.
.

. ~ - , .

wo92/ls67s PcT/uss2/ol~39 ` i? ~

There are many ways to generate diversity in a protein. At one extreme, we vary a few residues of the protein as much a~ possible (nFocused Mutagenesis"), e.g., we pick a set of five to seven residues and vary each through 13-20 possibilities. An alternative plan of mutagenesis (nDiffuse Mutagenesis") i5 to vary many more residues through a more limited set of choices (See VERS~6a and PAKU86). The variegation pattern adopted may fall between these extremes.
There is no fixed limit on the number of codons which can be mutated simultaneously. However, it is desirable to adopt a mutagenesis strategy which results in a reasonable probability that a possible PBD sequence is in fact displayed by at least one phage. Preferably, the probability that a mutein encoded by the vgDNA and composed of the least favored amino acids at each variegated position will be displayed by at least one independent transformant in the library is at least 0.50, and more preferably at least 0.90. (Muteins composed of more favored amino acids would of course be more likely to occur in the same library.) Preferably, the variegation is such as will cause a typical transformant population to di~play 106-107 different amino acid sequences by means of preferably not more than 10-fold more (more preferably not more than 3-fold) different DNA sequences.
For a mini-protein that lacks ~ helices and strands, one will, in any given round of mutation, preferably variegatP each of 4-6 non-cysteine codons so that they each encode at least eight of the 20 possible amino acids. The variegation at each codon could be customized to that position. Preferably, cysteine is not one of the potential substitutions, though it is not excluded.
3~ When the mini-protein is a metal finger protein, in a typical variegation strategy, the two Cys and two His .. . :, ' . .

W092/1~679 PCT/US92/01~39 ~ ~! ' 3~

residues, and optionally also the aforementioned Phe/Tyr, Phe and Leu residues, are held invariant and a plurality (usually 5-10) of the other residues are varied.
When the mini-protein is of the type featuring one S or more ~ helices and ~ strands, the set o~ potential amino acid modifications at any given position is picked to favor those which are less likely to disrupt the secondary structure at that position. Since the number of possibilities at each variable amino acid is more limited, the total number of variable amino acids may be greater without altering the sampling efficiency of the selection process.
For the last-mentioned class of mini-proteins, as well as domains other than mini-proteins, preferably not more than 20 and more preferably 5-10 codon~ will be variegated. However, if diffuse mutagenesis is employed, the number of codons which are variegated can be higher.
The decision as to which residues to modify is eased by knowledge of which residues lie on the surface of the domain and which are buried in the interior.
We choose residues in the PPBD to ~ary through consideration of several factors, including: a) the 3D
structure of the PPBD, b) sequences homologous to PPj3D, and c) modeling of the PP~D and mutants of the PP~D.
When the number of residues that could strongly influence binding without preventing the normal folding of the PP~D
is greater than the number that should be varied simultaneously, the user should pick a subset of those residues to vary at one time. The user picks trial levels of variegation and calculate the abundance~ of various sequences. The list of varied residues and the level of Yariegation a' each varied residue are adjusted until the composite variegation is commensurate with the sensitivity of the affinity separation and the number of independent transformants that can be made.

W O 92/15679 PC~r/~'S92/01539 2~ 0 .i ~ ~ ~J

Having picked which re~idues to vary, we now decide the range of amino acids to allow at each variable residue. The total level of variegation is the product of the number of variants at each varied residue. Each varied residue can have a different scheme of variegation, producing 2 to 20 different possibilities.
The set of amino acids which are potentially encoded by a given variegated codon are called its "substitution set~'.
The computer that controls a DNA ~ynthesizer, such as the Milligen 7500, can be programmed to synthesize any base of an oligo-nt with any distribution of nts by taking some nt substrates (e.g. nt phosphoramidites) from each of two or more reservoirs. Alternatively, nt substrates can be mixed in any ratios and placed in one of the extra reservoir for 80 called "dirty bottle"
synthesis. Each codon could be programmed differently.
The "mix" of bases at each nucleotide position of the codon determines the relative frequency of occurrence of the different amino acids encoded by that codon.
~ imply variegated codons are those in which those nucleotide positions which are degenerate are obtained from a mixture of two or more bases mixed in equimolar proportions. These mixtures are described in this specification by means of the standardized "am~iguous nucleotide~ code. In this code, for example, in the degenerate codon "SNT", "S" denotes an equimolar mixture of bases G and C, "N", an equimolar mixture of all four bases, and "T", the single invariant base thymidine.
Complexly variegated ~odons are those in which at least one of the three positions is filled by a base from an other than equimolar mixture of two of more bases.
Either simply or complexly variegated codons may be used to achieve the desired substitution set.
If we have no information indicating that a parti-cular amino acid or class of amino acid is appropriate, . .
.:

W092/1~679 PCT/US92/01~39 ~c~g,.
c~ t~

we strive to substitute all amino acids with equal probability because representation of one mini-protein above the detectable level i9 wasteful. Equal amounts of all four nts at each position in a codon (NMN) yields the amino acid distribution in which each amino acid is present in proportion to the number of codons that code for it. This distribution has the disadvantage of giving two basic residues for every acidic residue. In addition, 9iX times as much R, S, and L as W or M occur.
If five codons are synthesized with this distribution, each of the 243 sequences encoding some combination of L, R, and S are 7776-times more abundant than each of the 32 sequences encoding some combination of W and M. To have five Ws present at detectable levels, we must have each of the (L,R,S) sequences present in 7776-fold excess.
It i9 generally accepted that the sequence of amino acids in a protein or polypeptide determine the three-dimensional Rtructure of the molecule, including the possibility of no definite structure. Among polypeptides of definite length and sequence, some have a defined tertiary structure and most do not.
Particular amino acid residues can influence the tertiary structure of a defined polypeptide in several ways, including by:
a) affecting the flexibility of the polypeptide main chain, b) adding hydrophobic groups, c) adding charged groups, d) allowing hydrogen bonds, and e) forming cross-links, such as disulfides, chelation to metal ions, or bonding to prosthetic groups.
Flexibility:
GLY is the smallest amino acid, having two hydrogens attached to the Ca. Lecau~e GLY has no Cl, it confers the most flexibility on the main chain. Thus GLY occurs very w092/l5679 PCT/US9~/01~39 ` '3 ~

frequently in reverse turns, particularly in conjunction with PRO, ASP, ASN, SER, and THR.
The amino acids ALA, SER, CYS, ASP, ASN, LEU, MET, PHE, TYR, TRP, ARG, HIS, GLU, GLN, and LYS have unbranched ~ carbons. Of these, the side groups of SER, ASP, and ASN frequently make hydrogen bonds to the main chain and 90 can take on main-chain conformation~ that are energetically unfavorable for the others. VAL, ILE, and THR have branched B carbons which makes the extended main-chain conformation more favorable. Thus VAL and ILE
are most often seen in ~ ~heets. Because the side group of THR can easily form hydrogen bond~ to the main chain, it has less tendency to exist in a B sheet.
The main chain of proline is particularly constrained by She cyclic side group. The ~ angle i9 always close to -60. Most prolines are found near the surface of the protein.
~harge:
LYS and ARG carry a single positive charge at any pH
20 below 10.4 or 12.0, respectively. Nevertheless, the methylene groups, four and three respectively, of these amino acids are capable of hydrophobic interactions. The guanidinium group of ARG is capable of donating five hydrogens simultaneously, while the amino group of LYS
can donate only three. Furthermore, the geometries of these groups is quite different, RO that these groups are often not interchangeable.
ASP and GLU carry a single negative charge at any pH
above -4.5 and 4.6, respectively. Because ASP has but 3~ one methylene group, few hydrophobic interactions are possible. The geometry of ASP lends itself to forming hydrogen bonds to main-chain nitrogens which is consistent with ASP being found very often in reverse turns and at the beginning of helices. GLU is more often found in ~ helices and particularly in the amino-terminal portion of these helices because the negative charge of ,: , .

., .

WO92/15679 PCT/US92~01539 the side group has a stabilizing interaction with the helix dipole ~NICH88, SALI88).
HIS has an ionization pK in the physiological range, v z. 6.2. This pK can be altered by the proximity of charged groups or of hydrogen donators or acceptors. HIS
is capable of forming bonds to metal ions such as zinc, copper, and iron.
Hydrogen bonds:
Aside from the charged amino acids, SER, THR, ASN, GLN, TYR, and TRP can participate in hydrogen bonds.
Cr~ss links:
The most important form of cross link i8 the disul-fide bond formed between two thiols, especially the thiols of CYS residues. In a suitably oxidizing environ-ment, these bonds form spontaneously. These bonds cangreatly stabilize a particular conformation of a protein or mini-protein. When a mixture of oxidized and reduced thiol reagents are present, exchange reactions take place that allow the most stable conformation to predominate.
Concerning disulfides in proteins and peptides, see also ~ATZ90, M~TS89, PERR84, PERR86, SAUE86, WELL86, JANA89, HORV89, ~ISH85, and SCEN86.
Other cross links that form without need of specific enzymes include:
251) (CYS)4:Fe Rubredoxin (in CREI84, P.376) 2) (CYS) 4: Zn Aspartate Transcarbamylase (in CREI84, P.376~ and Zn-fingers (HARD90) 3 ) ( H I S ) 2 ( M E T ) ( C Y S ) : C u Azurin (in CREI84, P.376) and Basic "Blue" Cu Cucumber protein (GUSS88) 4) (HIS)4:Cu CuZn Yuperoxide dismutase 5) (CYS)4:(Fe4S~) Ferredoxin (in CREI84, P.376) 6) (CYS)2(HIS)2.Zn Zinc-fingers (GIBS88) 7j (CYS~3(HIS):Zn Zinc-fingers (GAUS87, GIBS88) W092/1~679 PCT1US92/01~39 ~ J~
4s Cross link~ having (HIS)2(MET)(CY~):Cu has the potential advantage that HIS and MET can not form other cross links without Cu.
Slmply Var~egated Codous The following simply variegated codon~ are useful because they encode a relatively balanced set of amino acids:
l) SNT which encodes the set [L,P,H,R,V,A,D,G]: a) one acidic (D) and one basic (R), b) both aliphatic (L,V) and aromatic hydrophobics (H), c) large (L,R,H) and small (G,A) side groups, d) ridged (P) and flexible (G) amino acids, e) each amino acid encoded once.
2) RNG which encodes the set [M,T,R,R,Y,A,E,G]: a) one acidic and two basic (not optimal, but acceptable), b) hydrophilics and hydrophobics, c) each amino acid encoded once.
3) RMG which encodes the set ~T,K,A,E]: a) one acidic, one basic, one neutral hydrophilic, b) three favor ~ helices, c) each amino acid encoded once.
4) VNT which encodes the ~et [L,P,H,R,I,T,N,S,V,A,D,G]:
a) one acidic, one basic, b) all classes: charged, neutral hydrophilic, hydrophobic, ridged and flex-ible, etc., c) each amino acid encoded once.
5) RRS which encodes the set [N,S,K,R,D,E,G~]: a) two acidic~, two basics, b) two neutral hydrophilics, c) only glycine encoded twice.
6) N N T w h i c h e n c o d e s t h e s e t ~F,S,Y,C,L,P,H,R,I,T,N,V,A,D,G]: a) sixteen DNA
sequences provide fifteen different amino acids;
only serine is repeated, all other~ are present in equal amounts (This allows very efficient sampling of the library.), b) there are equal numbers of acidic and basic amino acids (D and R, once each), c) all ma~or classes of amino acids are present:

.
. .
. .... ~ . .

W092/1~679 PCT/USs2/01539 acidic, basic, aliphatic hydrophobic, aromatic hydrophobic, and neutral hydrophilic.
7) N N G , w h i c h e n c o d e s t h e s e t ~L2,R2,S,W,P,Q,M,T,K,V,A,E,G, stop]: a) fair preponderance of residues that fa~or formation of ~-helices [L,M,A,Q,K,E; and, to a lesser extent, S,R,T]; b) encodes 13 different amino acids. (VHG
encodes a subset of the set encoded by NNG which encodes 9 amino acids in nine different DNA
sequences, with equal acids and bases, and 5/9 being helix-favoring.) For the initial variegation, NNT is preferred, in most cases. However, when the codon i9 encoding an amino acid to be incorporated into an ~ helix, NNG is preferred.
~ elow, we analyze several simple variegations as to the efficiency with which the libraries can be sampled.
Libraries of random hexapeptides encoded by (NNK) 6 have been reported (SCOT90, CWIR90). Table 130 shows the expected behavior of such libraries. NNX produces single codons for PHE, TYR, CYS, TRP, HIS, GLN, ILE, MET, ASN, LYS, ASP, and GLU (~ set); two codons for each of VAL, ALA, PRO, THR, and GLY (~ set); and three codons for each of LEU, ARG, and SER (Q set). We have separated the 64,000,000 possible sequences into 28 classes, shown in Table 130A, based on the number of amino acids from each of these ~ets. The largest class is ~n~ with -14.6 of the possible sequences. Aside from any selection, all the sequences in one class have the same probability of being produced. Table 13OB shows the probability that a given DNA sequence taken from the (NNX) 6 library will encode a hexapeptide belonging to one of the defined clas~es; note that only -6.3~ of DNA sequences belong to the ~Q~ class.

' ' W092/1~679 PCT/US9~/01~39 ~ ~ ~ ? ? 3 Table 130C shows the expected numbers of sequences in each class for libraries containing various numbers of independent transformants (v z. 1o6, 3-1o6, 107, 3-107, 10~, 3-10~, 109, and 3-109). At 1o6 independent transformants S (ITs), we expect to see s6~ of the nnQnQn class, but only 0.1~ of the ~ class. The vast majority of sequences seen come from classes for which less than 10~ of the class is sampled. Suppose a peptide from, for example, class ~ is isolated by fractionating the library for binding to a target. Consider how much we know about peptides that are related to the isolated sequence.
Because only 4% of the ~n~ class was sampled, we can not conclude that the amino acids from the ~ set are in fact the best from the Q set. We might have LEU at posi~ion 2, but ARG or SER could be better. Even if we isolate a peptide of the n~nnQ~ class, there is a notice-able chance that better members of the class were not present in the library.
With a library of 107 ITs, we see that ~everal classes have been completely sampled, but that the ~a~
class is only 1.1~ sampled. At 7.6 107 ITs, we expect display of 50~ of all amino-acid sequences, but the classes containing three or more amino acids of the ~ set are still poorly sampled. To achieve complete sampling of the (NNK) 6 library requires about 3-109 ITs, 10-fold larger than the largest (NNK) 6 library so far reported.
Table 131 shows expectations for a library encoded by (NNT) 4 (MNG)2 The expectations of abundance are independent of the order of the codons or of interspersed unvaried codons. This library encodes 0.133 times as many amino-acid sequences, but there are only 0.0165 times as many DNA sequences. Thus 5.0-107 ITs (i.e. 60-fold fewer than required for (NNX)6) gives almost complete sampling of the library. The resul~s would be slightly better for (NNT) 6 and slightly, but not much, worse for .. , . ~ - . . . ..

' . . .

W092/15679 PCT/US92/01~39 ~ ~ 48 (NNG)6. The controlling factor is the ratio of DNA
sequences to amino-acid sequences.
Table 132 shows the ratio of #DNA sequences/#AA
sequences for codons NNK, NNT, and NNG. For NNK and NNG, we have assumed that the PBD i8 displayed as part of an essential gene, such as gene III in Ff phage, as is indicated by the phrase "assuming stops vanish". It is not in any way required that such an essential gene be used. If a non-essential gene is used, the analysis would be slightly different; sampling of NNK and NNG
would be slightly less efficient. Note that (NNT)6 gives 3.6-fold more amino-acid sequences than (NNK) 5 but requires 1.7-fold fewer DNA sequences. Note also that (NNT)7 gives twice as many amino-acid sequences as (NNK)6, but 3.3-fold fewer DNA sequences.
Thus ! while it is possible to use a simple mixture (NNS, NNK or NNN) to obtain at a particular position all twenty amino acids, these simple mixtures lead to a highly biased set of encoded amino acids. This problem can be overcome by use of complexly variegated codons.
We first will present the mixture calculated (see W090/02809) to minimize the ratio of most favored amino acid to least favored amino acid when the nt distribution is subject to two constraints: equal abundances of acidic and basic amino acids and the least possible number of stop codons. We have simplified the search for an optimal nt distribution by limiting the third base to T or G (C or G is equivalent). However, it should be noted that the present invention embraces use of complexly variegated codons in which the third base is not limited to T or G (or to C or G).
The optimum distribution (the "fxS" codon) is shown in Table lOA and yields DNA molecules encoding each type amino acid with the abundances shown. Note that this chemistry encodes all twenty amino acids, with acidic and WO92/15679 PCT/US9~ 39 ? r3 ~?

basic amino acids being equiprobable, and the most favored amino acid (~erine) is encoded only 2.454 times as often as the least favored amino acid (tryptophan).
The "fxS" vg codon improves sampling most for peptides containing several of the amino acids ~F,Y,C,W,H-,Q,I,M,N,K,D,E] for which NNK or NNS provide only one codon. Its sampling advantages are most pronounced when the library is relatively small.
The results of searhing only for the complexly variegated codon which minimizes the ratio of most favored to least favored amino acid, without additional constraints, is shown in Table 10B. The changes are small, indicating that insisting on equality of acids and bases and minimizing stop codons costs us little. Also note that, without re~training the optimization, the prevalence of acidic and basic amino acids comes out fairly close. On the other hand, relaxing the restriction leaves a distribution in which the least favored amino acid is only .412 times as prevalent as SER.
The advantages of an NNT codon are discussed else-where in the present application. Unoptimized NNT
provides 15 amino acids encoded by only 16 DNA sequences.
It is possible to improve on NNT with the complexly variegated codon shown in Table 10C. This give~ five amino acids (SER, LEU, HIS, VAL, ASP) in very nearly equal amounts. A further eight amino acids (PHE, TYR, ILE, A~N, PRO, ALA, ARG, GLY) are present at 78~ the abundance of SER. THR and CYS remain at half the abun-dance of SER. When variegating DNA for disulfide-bonded mini-proteins, it is often de~lrable to reduce the prevalence of CY~. This distribution allows 13 amino acids to be seen at high level and give~ no stops; the optimized fxS di~tribution allows only 11 amino acids at high prevalence.

. . - , . ~ ~ , . , - -:
..

W092/15679 PCT~1S9~/01~39 ~j~3~

The NNG codon can also be optimized. When equimolar T,C,A,G are used in NNG, one obtains double doses of LEU
and ARG. Table lOD shows an approximately optimized MNG
codon. There are, under this variegation, four equally most favored amino acids: LEU, ARG, ALA, and GLU. Note that there is one acidic and one basic amino acid in this set. There are two equally least favored amino acids:
TRP and MET. The ratio of lfaa/mfaa i~ 0.5258. If this codon i9 repeated 5iX times, peptides composed entirely of TRP and MET are 2% as common as peptides composed entirely of the most favored amino acids. We refer to this a~ "the prevalence of (TRP/MET) 6 in optimized MNG6 vgDNA".
When synthesizing vgDNA by the lldirty bottle"
method, it is sometimes desirable to use only a limited number of mixes. One very useful mixture i9 called the "optimized NNS mixture" in which we average the first two positions of the fxS mixture: T, z 0.24, C1 = 0.17, A1 =
0.33, G~ = 0.26, the second position is identical to the first, C3 = G3 = 0.5. This distribution provides the amino acids ARG, SER, LEU, GLY, VAL, THR, ASN, and LYS at greater than 5% plus AhA, ASP, GLU, ILE, MET, and TYR at greater than 4~.
An additional complexly variegated codon is of interest. This codon is identical to the optimized NNT
codon at the first two positions and has T:G::90:lO at the third position. This codon provides thirteen amino acids (AhA, ILE, ARG, SER, ASP, LEU, VAL, PHE, ASN, GLY, PRO, TYR, and HIS) at more than 5.5~. THR at 4.3% and CYS at 3.9~ are more common than the LFAAs of NNK
(3.125%). The remaining five amino acids are present at less than l~. Thi3 codon has the feature that all amino acids are present; sequences having more than two of the low-abundance amino acids are rare. When we isolate an SBD using this codon, we can be reasonably sure that the WO92/15679 PCT/US92/01;39 ~ ~ Q 3 3 i' `~

first 13 amino acids were tested at each position. A
similar codon, based on optimized NNG, could be uqed.
Several of the preferred simple or complex variegated codons encode a ~et of amino acids which includes cysteine. This means that some of the encoded binding domains will feature one or more cysteines in addition to the invariant disulfide-bonded cysteines.
For example, at each NNT-encoded position, there is a one in sixteen chance of obtaining cysteine. If six codons are so varied, the fraction of domains containing additional cysteines is 0.33. Odd numbers of cysteines can lead to complications, see Perry and Wetzel (PERR84).
On the other hand, many disulfide-containing proteins contain cysteines that do not form disulfides, e.a.
trypsin. The possibility of unpaired cysteines can be dealt with in several wayf2:
Firqt, the variegated phage population can be passed over an immobilized reagent that strongly bindq free thiols, such as SulfoLink (catalogue number 44895 H from Pierce Chemical Company, Rockford, Illinois, 61105).
Another product from Pierce is TNB-Thiol Agaro~e (Cata-logue Code 20409 H). ~ioRad sells Affi-Gel 401 (catalogue 153-4599) for this purpose.
Second, one can use a variegation that excludes cysteines, such as:
NHT that gives [F,S,Y,L,P,H,I,T,N,V,A,D], VNS that gives [L2,p2,H,Q,R3,I,M,T2,N,K,S,V2,A2,E,D,G2], NNG that gives [L2,S,W,P,Q,R2,M,T,K,R,V,A,E,G,stop], 30 SNT that gives [L,P,H,R,V,A,D,G~, RNG that gives [M,T,X,R,V,A,E,G], RMG that give~ [T,K,A,E], VNT that gives ~,P,H,R,I,T,N,S,V,A,D,G], or RRS that gives [N,S,K,R,D,E,G2].
However, each of these schemes has one or more of the di~advantages, relative to NNT: a) fewer amino acids are . . ~ .
' , , ~ . , ' .. . . ..

W092/1~679 PCT/US9~/01~39 allowed, b) amino acid~ are not evenly provided, c) acidic and basic amino acids are not equally likely), or d) stop codons occur. Nonetheless, NNG, NHT, and VNT are almost as useful as NNT. NNG encodes 13 different amino S acids and one stop signal. Only two amino acids appear twice in the 16-fold mix.
Thirdly, one can enrich the population for binding to the preselected target, and evaluate selected sequences E~ hoc for extra cysteines. Those that contain more cysteines than the cysteines provided for conformational constraint may be perfectly usable. It is possible that a disulfide linkage other than the designed one will occur. This does not mean that the binding domain defined by the isolated DNA sequence is in any way unsuitable. The suitability of the isolated domains is best de~ermined by chemical and biochemical evaluation of chemically synthesized peptides.
Lastly, one can block free thiols with reagents, such as Ellman's reagent, iodoacetate, or methyl iodide, that specifically bind free thiols and that do not react with disulfides, and then leave the modified phage in the population. It is to be understood that the blocking agent may alter the binding properties of the mini-protein; thus, one might use a variety of blocking reagent in expectation that different binding domains will be found. The variegated population of thiol-blocked display phage are fractionated for binding. If the DNA sequence of the isolated binding mini-protein contains an odd number of cysteines, then synthetic means are used to prepare mini-proteins having each possible linkage and in which the odd thiol is appropriately blocked. Nishiuchi (NISH82, NISH86, and works cited therein) disclose methods of synthesizing peptides that contain a plurality of cysteines so that each thiol is protected with a different type of blocking group. These groups can be selecti~ely removed so that the disulfide pairing can be controlled. We envision using such a scheme with the alteration that one thiol either remains blocked, or is unblocked and then reblocked with a different reagent.
Use of NNT or NNG variegated codons leads to very efficient sampling of variegated libraries because the ratio of (different amino-acid sequences)/~different DNA
sequences) is much closer to unity than it is for NNK or even the optimized vg codon (fxS). Nevertheless, a few amino acids are omitted in each case. Both NNT and NNG
allow members of all important classes of amino acids:
hydrophobic, hydrophilic, acidic, basic, neutral hydrophilic, small, and large. After selecting a binding domain, a ~ubsequent variegation and selection may be desirable to achieve a higher affinity or specificity.
During this second variegation, amino acid possibilities overlooked by the preceding ~ariegation may be investigated.
In the second round of variegation, a preferred strategy i~ to vary each position through a new set of residues which includes the amino acid(s) which were found at that position in the successful binding domains, and which include as many as possible of the residues which were excluded in the first round of variegation.
Thus, later rounds of variegation test both amino acid positions not previously mutated, and amino acid substitutions at a previously mutated position which were not within the previous substitution set.
If the firQt round of variegation is entirely unsuccessful, a different pattern of variegation should be used. For example, if more than one interaction set can be defined within a domain, the residues varied in the next round of variegation should be from a different set than that probed in the initial variegation. If repeated failures are encountered, one may switch to a different IP~D.

. . :

WO 92/15679 PCT/US9~/01534 9"~Q ~ 3 NITY SELECTION OF TA~LGET-BINDING MI~TANTS
Affinity separation is used initially in the present invention to verify that the display system is working, e., that a chimeric outer surface protein has been expressed and transported to the surface of the phage and is oriented 80 that the inserted binding domain i8 accessible to target material. When used for this purpose, the binding domain is a known binding domain for a particular target and that target is the affinity molecule used in the affinity separation process. For example, a display system may be validated by using inserting DNA encoding BPTI into a gene encoding an outer surface protein of the phage of interest, and testing for binding to anhydrotrypsin, which is normally bound by BPTI.
If the phage bind to the target, then we have confirmation that the corresponding binding domain is indeed displayed by the phage. Phage which display the binding domain (and thereby bind the target) are separated from those which do not.
Once the display system is validated, it is possible to use a variegated population of phage which display a variety of different potential binding domains, and use affinity separation technology to determine how well they bind to one or more targets. This target need not be one bound by a known binding domain which is parental to the dicplayed binding domains, i.e., one may select for binding to a new target.
For example, one may variegate a ~PTI binding domain and test for binding, not to trypsin, but to another serine protease, such as human neutrophil elastase or cathepsin G, or even to a wholly unrelated target, such as hor~e heart myoglobin.
The term "affinity separation means~ includes, but i~ not limited to: a) affinity column chromatography, b) batch elution from an affinity matrix material, c) batch W092/1~679 PCT/~S9-/01~39 2 ~

elution from an affinity material attached to a plate, d) fluore~cence activated cell sorting, and e) electrophor-esis in the presence of target material. "Affinity material~' is used to mean a material with affinity for the material to be purified, called the ~analyten. In most cases, the association of the affinity material and the analyte is reversible 80 that the analyte can be freed from the affinity material once the impurities are washed away.
If affinity chromatography i9 to be u~ed, then:
1) the molecules of the target material must be of sufficient size and chemical reactivity to be applied to a solid support suitable for affinity separation, 2) after application to a matrix, the target material preferably does not react with water, 3) after application to a matrix, the target material preferably does not bind or degrade proteins in a non-specific way, and 4) the molecules of the target material must be suffi-ciently large that attaching the material to a matrix allows enough unaltered surface area (gener-ally at least 500 A2, excluding the atom that is connected to the linker) for protein binding.
Affinity chromatography is the preferred separation means, but FACS, electrophoresis, or other mean~ may also be used.
The present invention makes use of affinity separa-tion of phage to enrich a population for those phage carr~ing genes that code for proteins with desirable binding properties.
The present invention may be used to select for binding domains which bind to one or more target mater-ial~, and/or fail to bind to one or more target materials. Specificity, of course, is the ability of a binding molecule to bind strongly to a limited set of Wo92/1~679 PCTIUS92/Ot~9 `3 target materials, while binding more weakly or not at all to another set of target materials from which the first set must be distinguished.
Almost any molecule that is suitable for affinity separation may be used as a target. Possible targets include, but are not limited to peptides, soluble and insoluble proteins, nucleic acids, lipids, carbohydrates, other organic molecules (monomeric or polymeric), inorganic compounds, and organometallic compounds.
Serine proteases are an especially interesting class of potential target materials.
For chromatography, FACS, or electrophoresis there may be a need to covalently link the target material to a second chemical entity. For chromatography the second entity is a matrix, for FACS the second entity is a fluorescent dye, and for electrophoresis the second entity i9 a strongly charged molecule. In many cases, no coupling i9 required because the target material already has the desired property of: a) immobility, b) fluores-cence, or c) charge. In other cases, chemical orphysical coupling is required.
It is not necessary that the actual target material be used in preparing the immobilized or labeled analogue that is to be used in affinity separation; rather, suitable reactive analogues of the target material may be more convenient. Target material~ that do not have reactive functional groups may be immobilized by first creating a reactive functional group through the use of some powerful reagent, such as a halogen. In some cases, the reactive groups of the actual target material may occupy a part on the target molecule that is to be left undisturbed. In that case, additional functional groups may be introduced by synthetic chemistry.
Two very general methods of immobilization are widely used. The first i9 to biotinylate the compound of interest and then bind the biotinylated derivative to ..
.
:
, WO92/15679 PCT/US9'/01539 2~ nc~

immobilized avidin. The second method ~s to generate antibodies to the target material, immobilize the an~i-bodies by any of numerous methods, and then bind the target material to the immobilized antibodies. Use of antibodies is more appropriate for larger target materi-als; small targets (those comprising, for example, ten or fewer non-hydrogen atoms) may be 80 completely engulfed by an antibody that very little of the target i9 exposed in the target-antibody complex.
Non-covalent immobilization of hydrophobic molecules without resort to antibodies may also be used. A com-pound, such as 2,3,3-trimethyldecane is blended with a matrix precursor, such as sodium alginate, and the mixture is extruded into a hardening solution. The resulting beads will have 2,3,3-trimethyldecane dispersed throughout and exposed on the surface.
Other immobilization methods depend on the presence of particular chemical functionalities. A polypeptide will present -NH2 (N-t~rminal; Lysines), -COOH (C-ter-minal; Aspartic Acids; Glutamic Acids), -OH (Serines;
Threonines; Tyrosines), and -SH (Cysteines). A polysac-charide has free -OH groups, as does DNA, which has a sugar backbone.
The following table is a nonexhaustive review of reactive functional groups and potential immobilization reagents:

Group Reaaent R-NH2 Derivatives of 2,4,6-trinitro benzene sulfonates (TN~S), (CREI84, p.11) R-NH2 Carboxylic acid anhydrides, e.g.
derivatives of succinic anhydride, maleic anhydride, citraconic anhydride (CREI84, p.11) R-NH2 Aldehydes that form reducible Schiff ba9es tCREI84, p.12) guanido cyclohexanedione derivatives (CREI84, p.14) :
, ... . .

.

- : , .

WO92/1'679 PCT/US92/01~39 ''3 R-CO2H Diazo cmpds (CREI84, p.10) R-CO2- Epoxides (CREI84, p.10) 5 R-OH Carboxylic acid anhydrideQ
Aryl-OH Carboxylic acid anhydrides Indole ring ~enzyl halide and sulfenyl halides (CREI84, p.19) R-SH N-alkylmaleimides (CREI84, p.21) R-SH ethyleneimine derivative~ (CREI84, p.21) R-SH Aryl mercury compounds, (CREI84, P.21) R-SH Disulfide reagents, (CREI84, p.23) Thiol ethers Alkyl iodides, (CREI84, p.20) Ketones Make Schiff~s base and reduce with Na~H4. (CREI84, p.12-13) 25 Aldehydes Oxidize to COOH, ~ide supra.
R-SO3H Convert to R-SO2Cl and react with immobilized alcohol or amine.
30 R-PO3H Convert to R-PO2Cl and react with immobilized alcohol or am~ine.
CC double bonds Add HBr and then make amine or thiol.

The extensive literature on affinity chromatography and related techniques will provide furthe~ examples.
Matrices suitable for use as support materials include polystyrene, glass, agarose and other chromato-graphic ~upports, and may be fabricated into beads,sheets, columns, wells, and other forms as desired.
Suppliers of support material for affinity chromatography include: Applied Protein Technologies Cambridge, MA;
3io-Rad Laboratories, Rockville Center, NY; Pierce Chemical Company, Rockford, IL. Target materials are attached to the matrix in accord with the directions of the manufacturer of each matrix preparation with consideration of good presentation of the target.

. . - . -.

: . .: . ': . -, , : , , ~ '3~33 Early in the selection process, relatively high concentrations of target materials may be applied to the matrix to facilitate binding; target concentrations may subsequently be reduced to select for higher affinity SBDs.
The population of display phage is applied to an affinity matrix under conditions compatible with the intended use of the binding protein and the population is fractionated by passage of a gradient of Yome solute over the column. The process enriches for P~Ds having affinity for the target and for which the affinity for the target is least affected by the eluants used. The enriched fractions are those containing viable display phage that elute from the column at greater concentration of the eluant.
The eluants preferably are capable of weakening noncovalent interactions between the displayed PBDs and the immobilized target material. Preferably, the eluants do not kill the phage; the genetic message corresponding to successful mini-proteins is most conveniently amplified by reproducing the phage rather than by ia vitro procedures such as PCR. The list of potential eluants includes salts (including Na+, NH4+ , Rb+ , S04- -, H2P04-, citrate, K+, Li+, Cs+, HS04-, C03--, Ca++, Sr++, Cl-, P04---, HC03-, Mg++, Ba++, Br-, HP04- and acetate), acid, heat, compounds known to bind the target, and soluble target material (or analogues thereof).
Neutral solutes, such as etha~ol, acetone, ether, or urea, are frequently used in protein purification and are known to weaken non-covalent interactions between proteins and other molecules. Many of these species are, however, very harmful to bacteria and bacteriophage.
Urea ic known not to harm M13 up to 8 M. Salt is a preferred solute for gradient formation in most cases.
Decreasing pH is al~o a highly preferred eluant. In some cases, the preferred matrix is not stable to low pH so - ,.... . . :

, : ......... :
, Wog~/15679 PCT/US92/01539 ~Q`3 V`
,,. .

that salt and urea are the most preferred reagents.
Other solute~ that generally weaken non-covalent interac-tion between proteins and the target material of interest may also be used.
The uneluted display phage contain DNA encoding binding domains which have a sufficiently high affinity for the target material to resist the elution conditions.
The DNA encoding such successful binding domains may be recovered in a variety of ways. Preferably, the bound display phage are simply eluted by means of a change in the elution conditions. Alternatively, one may culture the phage in situ, or extract the target-containing matrix with phenol (or other suitable solvent) and amplify the DNA by PCR or by recombinant DNA techniques.
Or, if a site for a specific protease has been engineered into the display vector, the specific protease i9 used to cleave the binding domain from the GP.
Variation in the support material (polystyrene, glass, agarose, cellulose, etc.) in analysis of clones carrying SBDs is used to distinguish phage that bind to the support material rather than the target.
The harvested phage are now enriched for the binding-to-target phenotype by use of affinity separation involving the target material immobilized on an affinity matrix. Phage that fail to bind to the target material are washed away. It may be desirable to include a bacteriocidal agent, such as azide, in the buffer to prevent bacterial growth. The buffers used in chromatography include: a) any ions or other solutes needed to stabilize the target, and b) any ions or other solutes needed to stabilize the PBDs derived from the IPBD.
Recovery of phage that display binding to an affinity column is typically achieved by collecting fractions eluted from the column with a gradient of a chaotropic agent as de~cribed above, or of the target .
. . . -: ,' ~ ' ' ,, - . . .
.. .

W092/~S679 PCT/US92~01~39 J?

material in soluble form; fractions eluting later in the gradient are enriched for high-affinity phage. The eluted phage are then amplified in suitable host cells.
If some high-affinity phage cannot be eluted from the target in viable form, one may:
1) flood the matrix with a nutritive medium and grow the desired phage ~a situ, 2) remove parts of the matrix and use them to inoculate growth medium, 3) chemically or enzymatically degrade the linkage holding the target to the matrix so that GPs still bound to target are eluted, or 4) degrade the phage and recover DNA with phenol or other suitable solvent; the recovered DNA i9 used to transform cells that regenerate GPs.
It is possible to utilize combinations of these methods.
I~ should be remembered that what we want to recover from the affinity matrix is not the phage ~ç~ se, but the information in them as to the sequence of the successful epitope or binding domain.
AS described in W090/02809, one may modify the affinity separation of the method described to select a molecule that binds to material A but not to material ~, or that binds to both A and B at competing or noncompeting sites, or that do nQ~ bind to qelected targets .
SIJBSEQ~3NT PRODIJCTION
Using the method of the present invention, we can obtain a replicable phage that di~plays a novel protein domain having high affinity and specificity for a target material of interest. Such a phage carries both amino-acid embodiments of the binding protein domain and a DNA
embodiment of the gene encoding the novel binding domain.
The presence of the DNA facilitates expression of a protein comprising the novel binding protein domain : . . : -W092/15679 PCT/US92/~1539 within a high-level expression system, which need not be the same system used during the developmental process.
We can proceed to production of the novel binding protein in several ways, including: a) altering of the gene encoding the binding domain so that the binding domain i9 expressed as a soluble protein, not attached to a phage (either by deleting codons 5' of those encoding the binding domain or by inserting stop codons 3' of those encoding the binding domain), b) moving the DNA
encoding the binding domain into a known expression system, and c) utilizing the phage as a purification system. (If the domain is ~mall enough, it may be fea~ible to prepare it by conventional peptide synthesis methods.) l~ As previously mentioned, an advantage inhering from the use of a mini-protein as an IPBD is that it i9 likely that the derived SBD will also behave like a mini-protein and will be obtainable by means of chemical synthesis.
(The term "chemical synthesis", as used herein, includes the use of enzymatic agents in a cell-free environment.) Peptides may be chemically synthesized either in solution or on supports. Various combinations of stepwise synthesis and fragment condensation may be employed.
During synthesis, the amino acid side chains are protected to prevent branching. Several different protective groups are useful for the protection of the thiol groups of cysteines:
l) 4-methoxybenzyl (M~zl; Mob) (NISH82; ZAFA88), remov-able with HF;
2) acetamidomethyl (Acm)(NISH82; NISH86; ~ECR89c), removable with iodine; mercury ions (e.q., mercuric acetate); silver nitrate; and 3) S-para-methoxybenzyl.

.

: : - : ~

~. -, ~ .: ' ', WO9~/15679 PCT/~S92/Ot539 Other thiol protective groups may be found in standard reference works such as Greene, PROTECTIVE
GROUPS IN ORGANIC SYNTHESIS (1981).
Once the polypeptide chain has been synthesized, disulfide bonds must be formed. Possible oxidizing agents include air (NISH86), ferricyanide (NISH82), iodine (NISH82), and performic acid. Temperature, pH, solvent, and chaotropic chemicals may affect the course of the oxidation.
A large number of mini-proteins with a plurality of disulfide bonds have been chemically synthesized in biologically active form.
The successful binding domains of the present invention may, alone or as part of a larger protein, be used for any purpose for which binding proteins are suited, including isolation or detection of target materials. In furtherance of this purpose, the novel binding proteins may be coupled directly or indirectly, covalently o; noncovalently, to a label, carrier or support.
When used as a pharmaceutical, the novel binding proteins may be contained with suitable carriers or adjuvanants.
* * * * *
All references cited anywhere in this specification are incorporated by reference to the extent which they may be pertinent.

' , ::
,:
, ' ' ' , ~ , :' ~" '~ . ' , :

W092/~5679 PCT/US92/01539 All cells u~ed in the following examples are E~ coli cells .
EXAMPLE I
DISPI.AY OF ~PTI AS A ~SION TO M13 GENE VIII PROTEIN:
Example I involves display of ~3PTI on M13 as a fusion to the mature gene VIII coat protein. Each DNA
construction was confirmed by restriction digestion and DNA sequencing.
1. Construction of the viii-signal-sequence::b~ti::mature-viii-coat-protein Display Vector.
The operative cloning vectors are M13 and phagemids derived from M13. The initial construction was-in the fl-based phagemid pGEM-3Zf(-)~ (Promega Corp., Madison, WI.).
We constructed a gene encoding, in order,: i) a modified lacUVS promoter, ii) a Shine-Dalgarno sequence, iii) M13 gene VIII signal sequence, iv) mature BPTI, v) mature-M13-gene-VII~ coat protein, vi) multiple stop codons, and vii) a transcription terminator. This gene is illustrated in Table 102. The operator of ~a~ W 5 is the symmetrical lacO to allow tighter repression in the absence of IPTG. The longest segment that is identical to wild-type gene VIII is minimized so that genetic recombination with the co-existing gene VIII is unlikely.
1) OCV based upon pGEK-3Zf.
pGEM-3Zf~ (Promega Corp., Madison, WI.) is a vector containing the amp gene, bacterial origin of replication, bacteriophage fl origin of replication, a lacZ operon containing a multiple cloning site sequence, and the T7 and SP6 polymerase binding sequences.
a3mHI and SalI sites were introduced at the boundaries of the lacZ operon (to facilitate removal of the lacZ operon and its replacement with the synthetic gene~; this vector is named pGEM-MB3/4.
11) OCV ba~ed upon N13mp18.

.. . ~ - . . . - , . . .
.. : .. . .
. . . - . .: . . ., : : :-. . . . . .
. ., , : .
. ,- : . . :, .. .. .
.

W092/15679 PCTI~IS92/Ot~39 2t ~a3C~A,~

M13mpl8 (YANI85) i8 a vector (New England Biolabs, Beverly, MA.) consisting of the whole of the phage genome plus a 1~~ operon containing a multiple cloning site ~Mæss77). ~HI and SalI sites were introduced into M13mpl8 at the 5' and 3~ ends of the lacZ operon; this vector i9 named M13-MB1/2.
B) Synthetic Gene.
A synthetic gene (VIII-signal-~e~uence::mature-~si::mature-VIII-coat-~rotein) was con~tructed from 16 synthetic oligonucleotides, synthesized by Genetic Designs Inc. of Houston, Texas, via a method similar to those in KIMH89 and ASHM89. Table 102 contains an annotated version of this sequence. The oligonucleotides were phosphorylated, with the exception of the 5' most molecules, using standard methods, annealed and ligated in ~tages. The overhangs were filled in with T4 DNA
polymerase and the DNA was cloned into the HincII si~e of pGEM-3Zf(-); the initial construct is pGEM-M31. Double-stranded DNA of pGEM-MB1 was cut with PstI, filled in with T4 DNA polymerase and ligated to a SalI linker (New England BioLabs) so that the synthetic gene is bounded by ~HI and ~I sites (Table 102). The synthetic gene was obtained on a ~HI-SalI ca~sette and cloned into pGEM-MB3/4 and M13-MB1/2 using the introduced ~mHI and SalI
sites, to generate pGEM-MB16 and M13-MB15, respectively.
The synthetic insert was sequenced. The original Ribosome Binding Site (~BS) was in error (AGAGG instead of the designed AGGAGG) and we detected no expressed protein ln vivo and ln vitro.
C) Alterations to the ~ynthetic gene.
~) Riboso~e blndlng site (RBS).
In pGEM-MB16, a SacI-NheI DNA fragment (containing the R~S) was replaced with an oligonucleotide encoding a new RBS very similar to the RBS of E. coli phoA that is known to function.

,' ~ ' ; . ', WO92/15679 PCT~VS92/01;39 Original putative RBS (5'-to-3') GAGCTCagaggCTTACTATGAAGAAATCTCTGGTTCTTAAGGCTAGC
¦SacI¦ ¦ Nhe I ¦
New RBS (5'-to-3') GAGCTCTggaggaAATAAAATGAAGA~ATCTCTGGTTCTTAAGGCTAGC
¦SacI¦ ¦ Nhe I ¦
The putative R~3Ss are lower case and the initiating methionine codon is underscored and bold. The resulting construct is pGEM-~320. 1~ vitro expression of the gene carried by pGEM-MB20 produced a novel protein species of the expected size, about 14.5 kd.
1~) ac promoter.
To obtain higher expression of the fusion protein, the lac W S promoter was changed to a tac promoter. In pGEM-M~16, a ~HI-~p~II fragment (containing the lac W 5 promoter) was replaced with an oligonucleotide containing the -35 sequence of the trp promoter (Cf RUSS82) converting the lacW 5 promoter to tac. The vector is named pGEM-~322.
M~16 5'- GATCC tctagagtcggc TTTACA ctttatgcttc(cg-25 gctcg...... -3' 3'- G agatctcagccg aaatgt gaaatacgaag gc(cgagc..-5' ~3amHI

MB22 5'- GATCC actccccatccccctg TTGACA attaatcat -3 3'- G tgaggggtagggggac AACTGT taattagtagc-5 ~HI
(HpaII) Promoter and RBS variants of the fusion protein gene were con~tructed as follows:
Promoter RBS Encoded Protein.
pGEM-~316 lac old VIIIs.p.-BPTI-matureVIII
pGEM-~E320 lac new I' pGEM-~322 tac old '' 45 pGEM-MB26 tac new ~ ,.
, .' . ' ' ' ' .

W092/1~679 PCT/US92/Ot539 2 ~ 3~ ~

The synthetic genes from pGEM-MB20 and pGEM-MB26 were recloned into the altered phage vector M13-MB1/2 to generate the phage M13-M327 and M13-MB28 respectively.
I11. Slgnal Peptide Seguence.
5 1~ vitro expression of the ~ynthetic gene reguIated by tac and the "new" R~S produced a novel protein of the expected size for the unprocessed protein (-16 kd). In vivo expres~ion also produced novel protein of full size;
no processed protein could be seen on phage or in cell extracts by silver staining or by We~tern analysis with anti-BPTI antibody.
Thus we analyzed the signal sequence of the fusion.
Table 106 shows a number of typical signal sequences.
Charged residues are generally thought to be of great importance and are shown bold and underscored. Each signal sequence contains a long stretch of uncharged residues that are mostly hydrophobic; these are shown in lower case. At the right, in parentheses, is the length of the stretch of uncharged residues. We note that the fusions of gene VIII signal to 9PTI and gene III signal to BPTI have rather short uncharged segments. These short uncharged segments may reduce or prevent processing of the fusion peptides. We know that the gene Il~ signal sequence is capable of directing: a) insertion of the peptide comprising (mature-BPTI)::(mature-gene-III-protein) into the lipid bilayer, and b) translocation of ~PTI and most of the mature gene III protein across the lipid bilayer (vide infra). That the gene III remains anchored in the lipid bilayer until the phage is ~ssembled is directed by the uncharged anchor region near the carboxy terminus of the mature gene III protein (see Table 116) and not by the secretion signal sequence. The phoA signal sequence can direct secretion of mature BPTI
into the periplasm of E. coli (M~RK86). Furthermore, there is controversy over the mechanism by which mature W092/1~679 PCT/US92/01539 .~

authentic gene VIII protein comes to be in the lipid . bilayer prior to phage assembly.
Thus we replaced the DNA coding for the gene-VIII-putative-signal-sequence by each of DNA coding for: 1) the phoA signal sequence, 2) the bla signal sequence, and 3) the M13 gene III signal. Each of these replacements produces a tripartite gene encoding a fusion protein that comprises, in order: (a) a signal peptide that directs secretion into the periplasm of parts (b) and (c), derived from a first gene; (b) an initial potential binding domain (BPTI in this case), derived from a second gene (in this case, the second gene i~ an animal gene);
and (c) a structural packaging signal (the mature gene VIII coat protein), derived from a third gene.
The process by which the IPBD::packaging-signal fusion arrives on the phage surface i5 illustrated in Figure 1.
In Figure la, we see that authentic gene VIII protein appears (by whatever process) in the lipid bilayer 80 that both the amino and carboxy termini are in the cyto-plasm. Signal peptidase-I cleaves the gene VIII protein liberating the signal peptide (that is absorbed by the cell) and mature gene VIII coat protein that spans the lipid bilayer. Many copies of mature gene VIII coat protein accumulate in the lipid bilayer awaiting phage assembly (Figure lc). Some signal sequences are able to direct the translocation of quite large proteins across the lipid bilayer. If additional codons are inserted after the codons that encode the cleavage site of the signal peptidase-I of such a potent signal sequence, the encoded amino acids will be translocated across the lipid bilayer as shown in Figure lb. After cleavage by signal peptidase-I, the amino acids encoded by the added codons will be in the periplasm but anchored to the lipid bilayer by the mature gene ~III coat protein, Figure ld.
The circular single-stranded phage DNA is extruded through a part of the lipid bilayer containing a high . : ~ , . .
- ~ - .

~ . . ': ~ .
- . .
.

WO 9~ 6?9 PCI/US9~/01539 3 ~

c:oncentration of mature gene VIII coat protein; the carboxy terminus of each coat protein molecule packs near the DNA while the amino terminus packs on the outside.
Because the fusion protein is identical to mature gene 5 VIII coat protein within the trans-bilayer domain, the fusion protein will co-assemble with authentic mature gene VIII coat protein as shown in Figure le.
In each case, the mature VIII coat protein moiety is intended to co-assemble with authentic mature VIII coat
10 protein to produce phage particle having BPTI domains displayed on the surface. The source and character of the secretion signal sequence is not important because the signal sequence is cut away and degraded. The structural packaging signal, however, is quite important 15 because it must co-assemble with the authentic coat protein to make a working virus sheath.
a) Bacterlal Alkal~ne Phosphatase (~hoA) Slgnal Peptlde.
Construct pG~I-MB26 contains a SacI-AccIII frag~nent containing the new R13S and sequences encoding the 20 initiating methionine and the signal peptide of M13 gene VIII pro-protein. This fragment was replaced with a duplex (annealed from four oligo-nts) containing the RBS
and DNA coding for the initiating methionine and signal peptide of PhoA (INOU82) phage is pGEM-MB42. M13MB48 is 2~ a derivative of Ge~42. A ~HI-SalI DNA fragment from GenM~342, containing the gene construct, was ligated into a similarly cleaved vector M13MB1/2 giving rise to M13MB48.
PhoA RBS and signal peptide sequence WO92/1~679 PCT/US92/01~39 `J'~

5'-GAGCTCCATGGGAGAAAATAAA.ATG.AAA.CAA.AGC.ACG.-ISacIl met ly9 gln ser thr .ATC.GCA.CTC.TTA.CCG.TTA.CTG.TTT.ACC.CCT.GTG.ACA.-ile ala leu leu pro leu leu phe thr pro val thr .A~A.GCC.CGT.CCG.GAT.-3' lys ala arg pro asp......
IACCIII
b) ~-lactamase slgnal peptide.
To allow transfer of the ~-lactamase (amp) promoter and DNA coding for the signal peptide into the (mature-BPTI)::(mature-VIII-coat-protein) gene, we first introduced an AccIII site the amp gene adjacent to the codons for the ~-lactamase signal pept~de cleavage site (C~04-T and A~ol~G); vector is pGEM-MB40. We then ligated a ~mHI linker into the AatII site at nt 2260, 5I to the promoter; vector is pGEM-MB45. The ~HI-AccIII fragment now contains the am~ promoter, am~ RBS, initiating methionine and ~-lactamase signal peptide. This fragment was used to replace the corresponding fragment from pGEM-MB26 to generate construct pGEM-MB46.
25amp gene promoter and signal peptide sequences 5'-GGATCCGGTGGCACTTTTCGGGGA~ATGTGCGCGGAACCCCTATTTGTT-TATTTTTCTA~ATACATTCA~ATATGTATCCGCTCATGAGACAATAACC-CTGATA~ATGCTTCAATAATATTGAAAAAGGAAGAGT-ATG.AGT.ATT.CAA.CAT.TTC.CGT.GTC.GCC.CTT.ATT.CCC. m .TTT.-35met ser ile gln his phe arg val ala leu ile pro phe phe GCG.GCA.TTT.TGC.CTT.CCT.GTT.TTT.GCT.CAT.CCG.-3' ala ala phe cys leu pro val phe ala his pro ~) M13-gene-III-~ignal::bpti::mature-~III-coat-proteln We may also construct M13-~351 which would carry a ~mR
gene and a Ml3-qene-III-siqnal-peptide gene fragment fused to the previously described BPTI::mature-VIII-coat-45protein gene fragment. Because M13-M~351 contains no gene .., . ~ ..., , ... ~ -.
- : .

2 i ~ n ~

.III, the phage can not form plaques, but can render cells KmR. Infectiou~ phage particles can be obtained via helper phage. The gene III signal sequence is capable of directing (BPTI)::(mature-gene-III-protein) to the surface of phage (vide infra).
Summary of signal peptide fusion protein variants.
Signal Fusion Promoter R~3S sequence ~rotein 10 pGEM-M~26 tac new VIII BPTI/VIII-coat pGEM-MB42 tac new E~Qa BPTI/VIII-coat pGEM-M346 amp ~ am~ BPTI/VIII-coat pGEM-MB51 III III III BPTI/~III-coat (hypoth.) 15 M13 M~48 tac new hoA BPTI/VIII-coat 2. Analysis of the Protein Products Encoded by the SYnthetic (sianal-peptide::mature-bpti::viii-coat-~rotein) Genes 1) In vitro analy~l~
A coupled transcription/translation prokaryotic system (Amersham Corp., Arlington Heights, IL) was utilized for the _ vitro analysis of the protein products encoded by the BPTI/VIII synthetic gene and the derivatives.
Table 107 lists the protein products encoded by the listed vectors which are visualized by standard fluorography following in vitro synthesis in the presence of 35S-methionine and separation of the products using SDS
polyacrylamide gel electrophoresis. In each sample, a pre-~-lactamase product (-31 kd) can be seen. This is derived from the amp gene which is the common selection gene for the vectors. In addition, a (pre-BPTI/VIII) product encoded by the synthetic gene and variants can be seen a~ indicated. The migration of these species (-14.5 kd) is consi3tent with the expected size of the encoded proteins .
11) In vlvo analyQiR.

WO92/15679 PCT/US9~/01~39 ~ 72 The vectors detailed in sections (B) and (C) were freshly transfected into the E. coli strain XL1-blue ~Stratagene, La Jolla, CA) and in strain SEF'~ E. coli strain SE6004 (LISS85) carries the prlA4 mutation and i9 more permissive in secretion than strains that carry the wild-type prlA. SE6004 is F and is deleted for lacI; thus the cells can not be infected by M13 and lac W 5 and tac promoters can not be regulated with IPTG. Strain SEF' is derived from strain SE6004 (LISS85) by crossing with XL1-Blue~; the F' in XL1-Blue~ carries TcR and lacIq. SE6-004 is streptomycinR, Tcs while XL1-Blue~ is strepto-mycinS, TcR so that both parental strains can be killed with the combination of Tc and streptomycin. SEF' retains the secretion-permissive phenotype of the parental strain, SE6004(prlA4).
The fresh transfectants were grown in NZYCM medium (SAMB89) for 1 hour. IPTG was added over the range 1.0 ~M to 0.5 mM (to derepress lacW 5 and tac) and grown for an additional 1.5 hours.
Aliquots of cells expressing the synthetic-insert encoded proteins together with controls (no vector, moc~
vector, and no IPTG) were lysed in SDS gel loading buffer and electrophoresed in 20% polyacrylamide gels containing SDS and urea. Duplicate gels were either silver stained (Daiichi, Tokyo, Japan) or electrotransferred to a nylon matrix (~mmobilon from Millipore, ~edford, MA) for western analysis using rabbit anti-BPTI polyclonal antibodies.
Table 108 lists the interesting proteins seen by silver staining and western analysis of identical gels. We can see clearly by western analysis that IPTG-inducible protein species containing ~PTI epitopes exist in the test strains which are absent from the control strains.
In XL1-Blue~, the migration of this species is predomin-antly that of the unprocessed form of the pro-protein although a small proportion of the protein appears to .
, ~
. .
: - :

W092/1~679 PCT/US92/01~39 ~ ~ O .j~

migrate at a size consistent with that of a fully processed form. In SEF~, the processed form predominates, there being only a faint band corresponding to the unprocessed species.
Thus, in strain SEF', we ha~e produced a tripartite fusion protein that is specifically cleaved after the secretion signal sequence. We believe that the mature protein comprises BPTI followed by the gene VIII coat protein and that the coat protein moiety spans the membrane. One or more copies, perhaps hundreds of copies, of this protein will co-assemble into M13 derived phage or M13-like phagemids. This construction will allow us to a) mutagenize the BPTI domain, b) display each of the variants on the coat of one or more phage (one type per phage), and c) recover tho~e phage that display variants having novel binding properties with respect to target materials of our choice.
Rasched and Oberer (RASC86) report that phage produced in cells that express two alleles of gene ~III, that have differences within the first 11 residues of the mature coat protein, contain some of each protein. Thus, because we have achieved ln ~Q processing of the phoA(signal)::bpti::matureVIII fusion gene, it is highly likely that co-expression of this gene with wild-type VIII will lead to production of phage bearing ~PTI
domains on their surface. Mutagenesis of the E~i domain of these genes will provide a population of phage, each phage carrying a gene that codes for the variant of BPTI
displayed on the phage surface.
VIII Display Phage: Productlon, Preparatlon nd Analy~
i. Phage Production.
The OCV can be grown in X~1-Blue~ in the absence of IpTr-. Typically, a pla~ue plug is taken from a plate and grown in 2 ml of medium, containing freshly diluted cells, for 6 to 8 hours. Following centrifugation of this culture, the supernatant is titered. ThiQ is kept W092/15679 PcT/us92/ol339 as a phage stock for further infection, phage production, and display of the ~ene product of interest.
A 100-fold dilution of a fresh overnight culture of SEF' cells in 500 ml of NZCYM medium is allowed to grow to a cell density of 0.4 (Ab 600nm) in a shaker at 37C. To this culture is added a sufficient amount of the phage stock to give a MOI of 10 together with IPTG to give a final concentration of O.5 mM. The culture is allowed to grow for a further 2 hrs.
li. Phage Preparat~on and Purlficat~on.
The phage-producing bacterial culture is centrifuged to separate the phage in the supernatant from the bacterial pellet. To the supernatant is added one guarter, by volume, of phage precipitation solution (20~ PEG, 3.75 M
ammonium acetate) and PMSF to a final concentration of lmM. It is left on ice for 2 hours after which the precipitated phage is retrieved by centrifugation. The phage pellet is redissolved in TrisEDTA containing 0.1~
Sarkosyl and left at 4C for 1 hour after which any bacteria and bacterial debris is removed by centrifuga-tion. The phage in the supernatant is reprecipitated with PEG overnight at 4C. The phage pellet is re~uspended in LB medium and repreciptated another two times to remove the detergent. The phage is stored in LB
medium a~ 4C, titered, and used for analysis and binding studies .
A more stringent phage purification scheme involves centrifugation in a CsCl gradient (3.86 g of CsCl dissolved in NET buffer (0.1 M NaCl, lmM EDTA, O.lM Tris 30 pH 7.7) to make 10 ml). 1012 to 10l3 phage in TE Sarkosyl buffer are mixed with 5 ml of CsCl NET buffer and centrifuged overnight at 34K rpm in, for example, a Sorvall OTD-65B Ultracentrifuge. Aliquots of 400 ~1 are carefully removed. 5 ~1 aliqouts are removed from the fractions and analys~d by agarose gel electrophoresis after heating at 65C for lS minutes together with the ' : - , : .
' , .

W092/15679 PCT/US92/01~39 2 :~ 9 .~

gel loading buffer containing O.l~ SDS. Fractions containing phage are pooled, the phage reprecipitated and finally redissolved in LB medium to a concentration of 1012 to 1013 phage per ml.
S 111. Phage AnalyslQ.
The display phage are analyzed using standard methods of polyacrylamide gel electrophoresis and either silver staining of the gel or electrotransfer to a nylon matrix followed by analysis with anti-BPTI antiserum (Western analysis). Display of heterologous proteins is quantitated by comparison to serial dilutions of the starting protein, for example ~PTI, together with the display phage samples in the electrophoresis and Western analyses. An alternative method involves running a 2-fold serial dilution of a phage in which both the major coat protein and the fusion protein are silver stained.
Comparison of the ratios of the two protein species allows one to estimate the number of fusion proteins per phage since the number of VIII gene encoded proteins per phage 1-3000) is known.
Incorporation of fusion protein into bacteriophage.
In vivo expression of the processed BPTI:~III fusion protein, encoded by vectors GemMB42 and Ml3MB48, indicated that the processed fusion product is probably located within the cell membrane. Thus, it could be incorporated into the phage and that the BPTI moiety would be displayed at the phage surface.
SEF~ cells were infected with either Ml3M348 or Ml3mpl8, as control. The resulting phage were electrophoresed (-lO11 phage per lane) in a 20~ polyacrylamide gelcontaining urea followed by electrotransfer to a nylon matrix and western analy~is using anti-~3PTI rabbit serum.
A single species of protein was observed in phage derived from infection with the Ml3M348 stock phage which was not observed in the control infection. This protein migrated , W092/1567~ PCT/US9~/01539 ).J~

at an apparent size of -12 kd, consistent with that of the fully processed fusion protein.
Western analysis of SEF~ bacterial lysate with or without phage infection demonstrated another species of protein of about 20kd. This species was also present, to a lesser degree, in phage preparations which were simply PEG precipitated without further purification (for example, using nonionic detergent or by CsCl gradient centrifugation). A comparison of M13M348 phage preparations made in the presence or absence of detergent aldemonstrated that sarkosyl treatment and CsCl gradient purification did remove the bacterial contaminant while having no effect on the presence of the BPTI:VIII fusion protein. This indicates that the fusion protein has been incorporated and i9 a constituent of the phage body.
The time course of phage production and BPTI:VIII
incorporation was followed post-infection and after IPTG
induction. Phage production and fusion protein incorporation appeared to be maximal after two hours.
This time course was utilized in further phage productions and analyses.
Polyacrylamide electrophoresis of the phage prepara-tions, followed by silver staining, demonstrated that the preparations were essentially free of contaminating protein species and that an extra protein b~nd was present in M13MB48 derived phage which was not present in the control phage. The size of the new protein was consi~tent with that seen by we~tern analysis. A similar analysis of a serially diluted ~PTI:VIII incorporated phage demonstrated that the ratio of fusion protein to major coat protein was typically about 1:150. Since the phage contains about 3000 copies of the gene VIII
product, the phage population contains, on average, lO's of copies of fusion protein per phage.
Alteri~g the initiati~g methionlne of the natural gene VIII.

' ':

, ' ~ .

2 ~ 3 The OCV M13MB48 contains the ~ynthetic gene encoding the ~PTI:VIII fusion protein in the intergenic region of the ~odified M13mpl8 phage vector. The remainder of the ~rector consists of the M13 genome. To increase the phage incorporation of the fusion protein, we decided to diminish the production of the natural gene VIII product by altering the initiating methionine codon of this gene to CTG. In such cases, methionine is incorporated, but the rate of initiation is reduced. The change was achie~ed by site-specific oligonucleotide mutagenesis.
M K K S -rest of VIII
ACT.TCC.TC.ATG.AAA.AAG. TCT .
rest of XI - T S S stop Site-specific mutagenesis.
(L) K K S -rest of VIII
ACT.TCC.AG.CTG.AAA.AAG.TCT.
rest of XI - T S S stop Analyses of the phage derived from this modified vector indicated that there was a ~ignificant increase in the ratio of fusion protein to major coat protein.
Quantitative estimates indicated that within a phage population aR much as 100 copies of the BPTI :VIII fusion were incorporated per phage.
Dl~play of BPTI:VIII fusion protein by bacter~ophage.
The BPTI :VIII fusion protein had been shown to be incorporated into the body of the phage. This phage was analyzed further to demonstrate that the BPTI moiety was accessible to specific antibodies and hence displayed at the phage surface.
We added purified polyclonal rabbit anti-BPTI IgG to a known titer of phage. Following incubation, protein A-agarose beads are added to bind IgG and left to incubate overnight. The IgG-protein A beads and any bound phage are removed by centrifugation followed by a retitering of the supernatant to determine loss of phage. The phage WOg2/t~679 PCT/US92/01~39 ~ 78 hound to the beads can also be acid eluted and titered.
The assay includes controls, such as a W~ phage stock ~M13mpl8) and IgG purified from normal rabbit pre-immune ~erum.
Table 140 shows that while the titer of the WT phage is unaltered by anti-BPTI IgG, BPTI-IIIMK (positive control, ~ide infra), demonstrated a significant drop in titer with or without the extra addition of protein A beads.
(Note, since the BPTI moiety is part of gIIIp that binds phage to bacterial pili, this is expected.) Two batches of M13M~48 phage (containing the ~PTI:VIII fusion protein) demonstrated a significant reduction in titer, as judged by pfus, when anti-BPTI antibodies and protein A beads were added. The initial drop in titer with the antibody alone, differs somewhat between the two batches of phage. Retrieval of the immunoprecipitated phage, while not quantitative, was significant when compared to the WT phage control.
Further controls are shown in Table 141 and ~able 142.
The data demonstrated that the loss in titer observed for the BPTI:VIII containing phage is a result of the display of BPTI epitopes by these phage and the specific interaction with anti-BPTI antibodies. No significant interaction with either protein A agarose beads or IgG
purified from normal rabbit serum could be demonstrated.
The larger drop in titer for M13MB48 batch five reflects the higher level incorporation of the fusion protein in this preparation.
Functional~ty of the BPTI moiety in the BPTI-VIII dlsplay phage.
The previous two sections demonstrated that the BPTI:VIII fusion protein has been incorporated into the phage body and that the BPTI moiety i9 displayed at the phage ~urface. To demonstrate that the displayed molecule is functional, binding experiments were performed in a manner almost identical to that described . .
.' W092/15679 PCT/US9~/01~39 ~a~3 in the previou~ section except that proteases were used in place of antibodies. The display phage and controls are allowed to interact with immobilized proteases or immobilized inactivated proteases. Binding iB assessed by the 1099 in titer of the display phage or by determining the phage bound to the beads.
Table 143 shows the results of an experiment in which BPTI.VIII display phage, M13MB48, were allowed to bind to anhydrotrypsin-agarose beads. There was a significant drop in titer compared to WT phage (no displayed ~PTI).
A pool of phage (5AA Pool), each contain a variegated 5 amino acid èxtension at the BPTI:major coat protein interface, demonstrated a similar decline in titer. In control (table 143), very little non-specific binding of the display phage was observed with agarose beads carrying an unrelated protein (streptavidin).
Actual binding of the display phage is demonstrated by the data shown for two experiments in Table 144. The negative control is M13mpl8 and the positive control is BPTI-IIIMK, a phage in which the BPTI moiety, attached to the gene III protein, has been shown to be displayed and functional. M13MB48 and M13MB56 both bind to anhydrotryp in beads in a manner comparable to that of the positive control, being 40 to 60 times better than the negative control (non-display phage). Hence, functionality of the BPTI moiety, in the major coat fusion protein, was established.
Furthermore, Table 145 compares binding to active and inactivated trypsin by phage. The control phage, M13mpl8 and BPTI-III MR, demonstrated binding similar to that detailed elsewhere in the pre9ent application. Note that the relative binding is enhanced with trypsin due to the apparent marked reduction in the non-specific binding of the WT phage to the active protea9e. M13.3X7 and 35 M13.3X11, which each contain 'EGGGS' linker extensions at the domain interface, bound to anhydrotrypsin and trypsin WO92/15679 PCT/US92/OtS39 ~ ~ Q ~
~ 80 in a manner similar to BPTI-IIIMK phage. The binding, relative to non-display phage, was -100-fold higher in the anhydrotrypsin binding assay and at least l000-fold higher in the trypsin binding assay. The binding of another 'EGGGS~ linker variant (M13.3Xd) was similar to that of M13.3X7.
To demonstrate the specificity of binding the assays were repeated with human neutrophil elastase (HN~) beads and compared to that seen with tryp~in beads Table 146.
~PTI has a very high affinity for trypsin and a low affinity for HNE, hence the BPTI display phage should reflect these affinities when used in binding assays with these beads. The negative and positive controls for trypsin binding were as already described above while an additional positive control for the ENE beads, BPTI(K15L,MGNG)-III MA was included. The results, shown in Table 146, confirmed this prediction. M13M~48, M13.3X7 and M13.3Xll phage demonstrated good binding to trypsin, relative to WT phage and the HNE control (BPTI(K15L,MGNG)-III MA), being comparable to BPTI-IIIMK
phage. Conversely poor binding occurred when HNE beads were used, with the exception of the HNE positive control phage.
Taken together the accumulated data demonstrated that when BPTI is part of a fusion protein with the major coat protein of M13 phage, the molecule i9 both displayed at the surface of the phage and a significant proportion of it i9 functional in a specific protease binding manner.
* * *
EXAMP~E II
~ONSTR~CTION O~ BPTI/GENE-III DISPLAY VECTOR
DNA manipulations were conducted according to standard procedures as described in Maniatis et al. (MANI82) or Sambrook et al. (SAMB89). First the lacZ gene of M13-MBl/2 wa~ removed. M13-MBl/2 RF was cut with Bam~I and SalI and the large fragment was isolated. The recovered W092/t5679 PCT/US92/01539 2 i ~

~19 bp fragment was filled in with Klenow enzyme, ligated to a HindIII 8mer linker, and used to transfect XL1-Blue~ (Stratagene, La Jolla, CA) cells which were subsequently plated for plaque formation. RF DNA was prepared from chosen plaqueq and a clone, M13-MB1/2--delta, containing regenerated ~3~HI and SalI sites and a new HindIII site, all 500 bp upstream of the B~lII site (6935), was picked.
A unique NarI site was introduced into codons 17 and 18 of gene III (changing the amino acids from H-S to G-A, Cf. Table 110) in M13-MB1/2-delta:

P F Y S H S A E T
5'-ct ttc tat tct cac tcc gct gaa ac-3' wild-type 3'-ga aag ata aga ccg cgg cga ctt tg-5' mutagenizing oligo 5'-ct ttc tat tct ggc gcc gct gaa ac-3' mutant P F Y S G A A E T
The presence of a unique NarI site at nucleotide 1630 was confirmed by restriction enzyme analysis; the new vector is M13-MB1/2-delta-NarI. Phage MK was made by cloning the 1.3 Kb BamHI KmR fragment from plasmid pUC4K
(Pharmacia, Piscataway, NJ) into M13-MB1/2-delta-NarI.
Phage MK grows a~ well as wild-type M13, indicating that the changes at the cleavage site of gene III protein are not detectably deleterious to the phage.
INSERTION OF SYNT~ETIC BPTI GENE
The BPTI-III expression vector was constructed by standard means. The synthetic bpti-VIII fusion contains a NarI site that comprises the last two codons of the BPTI-encoding region. A second NarI site was introduced upstream of the ~PTI-encoding region by ligating the adaptor shown to AccIII-cut M13-MB26:
5'-TATTCTG~CGCCCGT -3' 3'-ATAAGACCGCGGGCAGGCC-5' ¦NarI¦ ¦ACCIII
The ligation sample was then restricted with NarI and a 180 bp DNA fragment encoding BPTI was isolated. RF DNA

., . ~ ' .
' - .
:

W092/lS679 PCT/US9~/01~39 of phage MK was digested with NarI, dephosphorylated, and ligated to the 180 bp fragment. Ligation samples were used to transfect XL1-Blue~ which were plated for Km~
plaques. DNA, isolated from phage derived from plaques was test for hybridization to a 32P-phosphorylated double stranded DNA probe corresponding to the BPTI gene. Large scale RF preparations were made for clones exhibiting a strong hybridization signal. Restriction enzyme digestion analysis confirmed the insertion of a single copy of the synthetic BPTI gene into gene III of MK to generate phage MK-BPTI. Subsequent DNA sequencing con-firmed that the sequence of the b~ti-III fusion gene is correct and that the correct reading frame is maintained.
Table 116 shows the entire coding region, the translation into protein sequence, and the functional parts of the polypeptide chain.
~XPRESSION OF TaE 8PTI-III P~SION GENE IN VITRO
MK-BPTI RF DNA was added to a coupled prokaryotic transcription-translation extract (Amersham). Newly synthesized radiolabelled proteins were produced and ~ubsequently separated by electrophoreRis on a 15~ SDS-polyacrylamide gel. The MX-BPTI D~A directs the synthecis of an unprocessed gene III fusion protein which is 7 Kd larger than the WT gene III, consistent with insertion of 58 amino acids of BPTI into gene III
protein. We immunoprecipitated radiolabelled proteins from the cell-free prokaryotic extract. Neither rabbit anti(M13-gene-VIII-protein) IgG nor normal rabbit IgG
were able to immunoprecipitate the gene III protein encoded by either MK or MX-BPTI. However, rabbit anti-BPTI IgG is able to precipitate the gene III protein encoded by MR-BPTI but not by MK. Thig confirms that the increase in size of the III protein encoded by MX-BPTI is attributable to the insertion of the BPTI protein.
W~STER~ ANALYSIS

- ~ . , .
. .
... . . .

WO92/1~679 PCT/US92/01539 ~l ~.3 ~j ,9~

Phage were recovered from cultures by PEG precipitation.
To remove residual cells, recovered phage were resuspended in a high salt buffer and centrifuged, as per instructions for the MUTA-GENE~) M13 ln vitro Mutagenesis Kit (Catalogue Number 170-3571, Bio-Rad, Richmond, CA).
Aliquots of phage (containing up to 40 ~g of protein) were electrophoresed on a 12.5% SDS-urea-polyacrylamide gel and protein~ were electro-tranqferred to a sheet of Immobilon. Western blots were developed using rabbit anti-BPTI serum, which had previously been incubated with an E. coli extract, followed by goat ant-rabbit antibody conjugated to alkaline phosphatase. An immunoreactive protein of 67 Kd is detected in preparations of the MK-~PTI but not the MK phage. The size of the immunore-active protein is consistent with the predicted size of a processed BPTI-III fusion protein (6.4 Kd plus 60 Kd).
These data indicate that BPTI-6pecific epitopes are presented on the surface of the MK-BPTI phage but not the MX phage.
~u-lRALIZA~ION OF P~aGE TITER WIT~ AGAROSE-rMMOBI~IZED

Anhydro-trypsin is a derivative of trypsin having the active site serine converted to dehydroalanine. Anhydro-trypsin retains the specific binding of trypsin but not25 the protease activity. Unlike polyclonalantibodies, anhydro-trypsin is not expected to bind unfolded BPTI or incomplete fragments.
Phage MX-BPTI and MK were diluted to a concentration 1.4-10l2 particles per ml. in TBS buffer (PARM88~ contain-ing 1.0 mg/ml ~SA. 30 ~1 of diluted phage were added to 2, 5, or 10 ~1 of a 50~ slurry of agarose-immobilized anhydro-trypsin (Pierce Chemical Co., Rockford, IL) in TBS/BSA buffer. Following incubation at 25C, aliquots were removed, diluted in ice cold LB broth and titered for plaque-forming units on a lawn of XLl-Blue~ cells.
Table 114 shows that incubation of the MK-BPTI phage with . . .
- :

- : . . .

W0~2/15679 PCT/VS9~/OlS39 immobilized anhydro-trypsin results in a very significant loss in titer over a four hour period while no such effect is observed with the MK (control) phage. The reduction in phage titer is also proportional to the amount of immobilized anhydro-trypsin added to the MK-BPTI phage. Incubation with 5 ~1 of a 50~ slurry of agarose-immobilized streptavidin (Sigma, St. Louis, MO) in TBS/BSA buffer does not reduce the titer of either the MK-BPTI or MK phage. These data are consistent with the presentation of a correctly-folded, functional BPTI
protein on the surface of the MX-BPTI phage but not on the MX phage. Unfolded or incomplete ~PTI domains are not expected to bind anhydro-trypsin. Furthermore, unfolded BPTI domains are expected to be non-specifically sticky.
= IZATION OF P~AGE TITE~ WIT~ ANTI-BPTI ANTIBODY
MX-BPTI and MK phage were diluted to a concentration of 4-103 plaque-forming units per ml in LB broth. 15 ~1 of diluted phage were added to an equivalent volume of either rabbit anti-BPTI serum or normal rabbit serum (both diluted 10-fold in LB broth). Following incubation at 37C, aliquots were removed, diluted by 104 in ice-cold LB broth and titered for pfus on a lawn of XL1-Blue~.
Incubation of the MK-BPTI phage with anti-BPTI serum results in a steady loss in titer over a two hour period while no such effect is observed with the MX phage. As expected, normal rabbit ~erum does not reduce the titer of either the MK-BPTI or the MX phage. Prior incubation of the anti-BPTI ~erum with authentic BPTI protein but not with an equivalent amount of E. coli protein, blocks the ability of the serum to reduce the titer of the MX-BPTI phage. These data are consistent with the presentation of BPTI-specific epitopes on the surface of the MX-BPTI phage but not the MK phage. More specifi-cally, the data indicates that these BPTI epitopes areassociated with the gene III protein and that a~sociation , - .

,, .
.

WO92/15679 PCT/US9~/Ot~39 2 ~

of this fusion protein with an anti-BPTI antibody blocks its ability to mediate the infection of cells.
N~TRALIZATION OF P~AGE TITER WIT~ TRYPSIN

MX-BPTI and MX phage were diluted to a concentration of 5 4~108 plaque-forming units per ml in LB broth. Diluted phage were added to an equivalent volume of trypsin diluted to various concentrations in LB broth. Following incubation at 37C, aliquots were removed, diluted by 104 in ice cold LB broth and titered for plaque-for,ming units on a lawn of XLl-Blue~M). Incubation of the MX-BPTI
phage with 0.15 ~g of trypsin results in a 70~ loss in titer after two hours while only a 15~ 1099 in titer is observed for MK phage. A reduction in the amount of trypsin added to phage results in a reduction in the 1098 of titer. However, at all trypsin concentrations inves-tigated , the MK-BPTI phage are more ~ensitive to incuba-tion with trypsin than the MK phage. Thus, association of the BPTI-III fusion protein displayed on the surface of the MX-BPTI phage with trypsin blocks its ability to mediate the infection of cells.
The reduction in titer of phage MX by tryp~in is an example of a phenomenon that is probably general:
proteases, if present in sufficient quantity, will degrade proteins on the phage and reduce infectivity.
~,,~,,,~
~m'"
AFFINITY SE~ECTION SYSTEM
Affinity Selection with Immobilized A~hydro-Trypsin MX-BPTI and MX phage were diluted to a concentration of 1.4-10~2 particles per ml in TBS buffer (PARM88) containing 1.0 mg/ml BSA. We added 4.0-101 phage to 5 ~1 of a 50% slurr,y of either agarose-immobilized anhydro-trypsin beads (Pierce Chemical Co.) or agarose-immobili7ed streptavidin beads (Sigma) in TBS/BSA.
Following a 3 hour incubation at room temperature, the bead~ were pelleted by centrifugation for 30 seconds at .
, . ~ .. . . -, - . . ' .
. . , ~ , . . ~ . .
, W092/1~679 PCT/US92/01539 ~ 86 5000 rpm in a microfuge and the supernatant fraction was collected. The beads were washed 5 time~ with T3S/Tween buffer (PARM88) and after each wash the beads were pelleted by centrifugation and the supernatant was removed. Finally, beads were resuspen~ed in elution buffer (0.1 N HCl containing 1.0 mg~ml BSA adjusted to pH
2.2 with glycine) and following a 5 minute incubation at room temperature, the beads were pelleted by centrifuga-tion. The supernatant was removed and neutralized by the addition of 1.0 M Tris-HCl buffer, pH 8Ø
Aliquots of phage samples were applied to a Nytran membrane using a Schleicher and Schuell (Keene, NH) filtration minifold and phage DNA was immobilized onto the Nytran by baking at 80C for 2 hours. The baked filter was incubated at 420C for 1 hour in pre-wash solution (MANI82) and pre-hybridization solution (5Prime-3Prime, West Chester, PA). The 1.0 Kb Na~I (base 1630)/XmnI tbase 2646) DNA fragment from MK RF was radioactively labelled with 32P-dCTP using an oligolabell-ing kit (Pharmacia, Piscataway, NJ). The radioactiveprobe was added to the Nytran filter in hybridization solution (5Prime-3Prime) and, following overnight incuba-tion at 42C, the filter was washed and autoradiographed.
The efficiency of this affinity selection system can be semi-quantitatively determined using a dot-blot procedure. Exposure of MK-~PTI-phage-treated anhydro-trypsin beads to elution buffer releases bound MX-BPTI
phage. Streptavidin beads do not retain phage MK-BPTI.
Anhydro-trypsin beads do not retain phage MR. In the 30 experiment depicted in Table 115, we estimate that 20~ of the total MK-BPTI phage were bound to 5 ~1 of the immobilized anhydro-trypsin and were subse~uently recovered by washing the beads with elution buffer (pH
2.2 HCl/glycine). Under the same conditions, no detectable MK-BPTI phage were bound and 8ubsequently recovered from the streptavidin beads. The amount of MK-- : - .
.. . . .
.

W092/15679 PCT/US92/01~39 2~ ?

~PTI phage recovered in the elution fraction is proportional to the amount of immobilized anhydro-trypsin added to the phage. No detectable MK phage were bound to either the immobilized anhydro-trypsin or streptavidin beads and no phage were recovered with elution buffer.
These data indicate that the affinity selection system described above can be utilized to select for phage displaying a specific folded protein (in this case, BPTI). Unfolded or incomplete ~PTI domains are not expected to bind anhydro-trypsin.
Affinity Selection wlth Anti-3PTI antibodles MK-BPTI and MK phage were diluted to a concentration of 1-101 particles per ml in Tris buffered saline solution (PARM88) containing 1.0 mg/ml ~SA. Two 10~ phage were added to 2.5 ~g of either biotinylated rabbit anti-BPTI
IgG in TBS/BSA or biotinylated rabbit anti-mouse antibody IgG (Sigma) in TBS/BSA, and incubated overnight at 4C.
A 50~ slurry of streptavidin-agarose (Sigma), wa~hed three times with TBS buffer prior to incubation with 30 mg/ml BSA in TBS buffer for 60 minutes at room tempera-ture, was washed three times with TBS/Tween buffer (PARM8~) and resuspended to a final concentration of 50~
in this buffer. Samples containing phage and biotinylated IgG were diluted with TBS/Tween prior to the addition of streptavidin-agarose in TBS/Tween buffer.
Following a 60 minute incubation at room temper~ture, streptavidin-agarose beads were pelleted by centrifu-gation for 30 seconds and the supernatant fraction was collected. The beads were washed 5 times with T~S/Tween buffer and after each wash, the beads were pelleted by centrifugation and the supernatant was removed. Finally, the streptavidin-agarose beads were resuspended in elution buffer (0.1 N HCl containing 1.0 mg/ml BSA
adjusted to pH 2.2 with glycine), incubated 5 minute at room temperature, and pelleted by centrifugation. The : :: . .: . ..
- . ~ - , : .
. ~ - .
: . , : . ' -.
-.. . .

W09~/15679 PCT/US9'/01~39 ~ 88 supernatant was removed and neutralized by the additionof 1.0 M Tris-HCl buffer, pH 8Ø
Aliquots of phage samples were applied to a Nytran membrane using a Schleicker and Schuell minifold appar-atus. Phage DNA was immobilized onto the Nytran bybaking at 80C for 2 hours. Filters were washed for 60 minutes in pre-wash solution (MANI82) at 42C then incubated at 42C for 60 minutes in Southern pre-hybri-dization solution (5Prime-3Prime). The 1.0 Kb NarI
(1630bp)/XmnI (2646 bp) DNA fragment from MK RF was radioactively labelled with 32P-~dCTP using an oligo-labelling kit (Pharmacia, Piscataway, NJ). Nytran membranes were transferred from pre-hybridization solution to Southern hybridization solution (5Prime--3Prime) at 42C. The radioactive probe was added to the hybridization solution and following overnight incubation at 42C, the filter was washed 3 times with 2 x SSC, 0.1 SDS at room temperature and once at 65C in 2 x SSC, 0.1 SDS. Nytran membranes were subjected to autoradiography.
The efficiency of the affinity selection system can be semi-quantitatively determined using the above dot blot procedure. Comparison of dots Al and B1 or Cl and Dl indicates that the majority of phage did not stick to the streptavidin-agarose beads. Washing with TBS/Tween buffer removes the majority of phage which are non--specifically associated with streptavidin beads.
Exposure of the streptavidin beads to elution buffer releases bound phage only in the case of MK-BPTI phage which have previously been incubated with biotinylated rabbit anti-BPTI IgG. This data indicates that the affinity selection cystem described above can be utilized to select for phage displaying a specific antigen (in this case BPTI). We estimate an enrichment factor of at lea3t 40 fold based on the calculation Percent MK-BPTI phage recovered Enrichment Factor =

.

, .

W092/15679 PCT/US9'/01~39 2 ~

.

Percent MK phage recovered EXAMPLE III
5 BPTI :VIII 90~JNDARY EXT~SIONS .
To increase the flexibility between BPTI and mature gVIIIp, we introduced codons for peptide extensions between these domains.
Adding block extension~ to the fusion ~rotein interface.
The M13 gene III product contains Istalk-like~ regions as implied by electron micrographic visualization of the bacteriophage (LOPEa5). The predicted amino acid sequence of this protein contains repeating motifs, which include:
glu.gly.gly.gly.ser (EGGGS) seven times gly.gly.gly.ser ~GGGS) three times glu.gly.gly.gly.thr (EGGGT) once.
The aim of this section was to insert, at the domain interface, multiple unit extensions which would mirror the repeating motifs observed in the III gene product.
Two synthetic oligonucleotides were synthesized. We picked the third base of these codons so that translation of the oligonucleotide in the opposite direction would yield SER. When annealed the synthetic oligonucleotides give the following unit duplex sequence (an EGGGS
linker):
E G G G S
5 ' C.GAG.GGA.GGA.GGA.TC 3' 3 I TC . CCT . CCT . CCT.AG(; . C 5 ' (L) (S) (S) (S) (G) The duplex has a common two base pair 5' o~erhang (GC) at either end of the linker which allows for both the ligation of multiple units and the ability to clone into the unique NarI recognition sequence pxesent in OCV's M13MB48 and Gem M~42. This site is positioned within 1 codon of the DNA encoding the interface. The cloning of an EGGGS linker (or multiple lin~er) into the ~ector NarI

. . . : , , . . . ~ ' . : . , : .
, WO92/1~679 PCT/US92/01~39 ~ 393 site destroys this recognition sequence. Insertion of the EGGGS linker in reverse orientation leads to insertion of GSSSL into the fusion protein.
Addition of a single EGGGS linker at the NarI site of the gene shown in Table 113 leads to the following gene:
79 80 80a 80b 80c 80d 80e 81 82 83 84 G G E G G G S A A E G
GGT.GGC.GAG.GGA.GGA.GGA.TCC.GCC.GCT.GAA.G&T

Note that there is no preselection for the orientation of the linker(s) inserted into the OCV and that multiple linkers of either orientation (with the predicted EGGGS
or GSSSL amino acid sequence) or a mixture of orientations (inverted repeats of DNA) could occur.
A ladder of increasingly large multiple linkers was established by annealing and ligating the two starting oligonucleotides containing different proportions of 5' phosphorylated and non-phosphorylated ends. The logic behind this is that ligation proceeds from the 3' unphos-phorylated end of an oligonucleotide to the 5' phosphor-ylated end of another. The use of a mixture of phosphor-ylated and non-phosphorylated oligonucleotides allows for an element of control over the extent of multiple linker formation. A ladder showing a range of insert sizes was readily detected by agarose gel electrophoresis spanning 15 bp (1 unit duplex-5 amino acids) to greater than 600 base pairs (40 ligated linkers-200 amino acids).
Large inverted repeats can lead to genetic instability.
Thus we chose to remove them, prior to ligation into the OCV, by digesting the population of multiple linkers with the restriction enzymes AccIII or XhoI, since the linkers, when ligated 'head-to-head' or 'tail-to-taill, generate these recognition sequences. Such a digestion significantly reduces the range in sizes of the multiple linkers to between 1 and 8 linker units (i.e. between 5 .. ... .. . . ~ . - : , .

.

WO92t15679 PCT/US92/01539 and 40 amino acids in steps of 5), as assessed by agarose gel electrophoresis.
The linkers were ligated (as a pool of different insert sizes or as gel-purified discrete fragments) into NarI
cleaved OCVs M13MB48 or GemMB42 using standard method~.
Following ligation the restriction enzyme NarI was added to remove the self-ligating starting OCV (since linker insertion destroys the ~I recognition sequence). This mixture was used to transform XL-1 blue cells and appro-priately plated for plaques (OCV M13MB48) or ampicillin resistant colonies (OCV GemMB42).
The transformants were screened using dot blot DNA
analysis with one of two 32p labeled oligonucleotide probes. One probe consisted of a sequence complementary to the DNA encoding the P1 loop of BPTI while the second had a sequence complementary to the DNA encoding the domain interface region. Suitable linker candidates would probe positively with the first probe and negatively or poorly with the second. Plaque purified clones were used to generate phage stocks for binding analyses and BPTI display while the Rf DNA~derived from phage infected cells was used for restriction enzyme analysis and sequencing. Representative insert sequences of selected clones analyzed are as follow~:
M13.3X4 tGG)C.GGA.TCC.TCC.TCC.CT(C.GCC~) gly ser ser ser leu M13.3~7 (G C.GAG.GGA.GGA.GGA.TC~C.GCC) glu gly gly gly ser M13.3X11 (GG)C.GAG.GGA.GGA.GGA.TCC.GGA.TCC.TCC.
glu gly gly gly ser gly ser ser TCC.CTC.GGA.TCC.TCC.TCC.CT~C.GCCC) ~er leu gly ser ser ser leu These highly flexible oligomeric linkers are believed to be useful in joining a binding domain to the major coat ~gene VIII) protein of filamentous phage to facilitate .
, ,, , :
.

W092/1~679 PCT/US92/01s39 the display of the binding domain on the phage surface.
They may also be useful in the construction o~ chimeric OSPs for other genetic packages as well.

S Incorporation of interdomain extension fuslon proteinR
into phage.
A phage pool containing a variegated pentapeptide extension at the ~PTI:coat protein interface wa~ used to infect SEF' cells. Using the criteria of the previous section, we determined that extended fusion proteins were incorporated into phage. Gel electrophoresis of the generated phage, followed by silver staining or western analysis with anti-BPTI rabbit serum, demonstrated fusion proteins that migrated similarly to, but discernably slower than, the starting fusion protein.
With regard to the 'EGGGS linker' extensions of the domain interface, individual phage stocks predicted to contain one or more 5-amino-acid unit extensions were analyzed in a similar fashion. The migration of the extended fusion proteins were readily distinguishable from the parent fusion protein when viewed by western analysis or silver staining. Those clones analyzed in more detail included M13.3X4 (which contains a ~ingle inverted EGGGS linker with a predicted amino acid sequence of GSSSL), M13.3X7 (which contains a correctly orientated linker with a predicted amino acid sequence of EGGGS), M13.3Xll (which contains 3 linkers with an inversion and a predicted amino acid sequence for the extension of EGGGSGSSSLGSSSL) and M13.3Xd which contains an extension consisting of at least 5 linkers or 25 amino acids.
The extended fusion proteins were all incorporated into phage at high levels (on average lO's of copies per phage were present and when analyzed by gel electrophoresis migrated rates consistent with the predicted size of the extension. Clones M13.3X4 and M13.3X7 migrated at a . ~ :

W092/lS679 PCT/US92/Ot539 21~

position very similar to but discernably different from the parent fusion protein, while M13.3Xll and M13.3Xd w e r e m a r k e d 1 y 1 a r g e r EXAMPLE IV
Peptide phage The following materials and methods were used in the examples which follow.
1. Peptide Phage HPQ6, a putative disulfide-bonded mini-protein, was displayed on M13 phage as an insert in the gene III
protein (gIIIp). M13 has about five copies of gIIIp per virion. The phage were constructed by standard methods.
HPQ6 includes the sequence CHPQFPRC characteristic of Devlin's streptavidin-binding E peptide (DEV~90), as well as a F.X, recognition site (see Table 820). HPQ6 phage were shown to bind to streptavidin.
An unrelated display phage with no affinity for streptavidin, MKTN, was used as a control.
2. Streptavidin.
Commercially available immobilized to agarose beads (Pierce). Streptavidin (StrAv) immobilized to 6% beaded agarose at a concentration of 1 to 2 mg per ml gel, provided as a 50% slurry. Also available as free protein (Pierce) with a specific activity of 14.6 units per mg (1 unit will bind 1 ~g of biotin). A stock solution of 1 mg per ml in PBS containing O.01% azide is made.
3. D-Blotin.
Commercially available (Boehringer Mannheim) in crystallized form. A stock solution of 4 mM is made.
4. Streptav~din coating of microtiter well plates.
Immulon (#2 or #4) strips or plates are used. lOO~L of StrAv stock is added to each 250 ~L capacity well and incubated overnight at 4C. The stock is removed and . : . : . . , . :
..
, - . . . . .

.
.

WOg2/1~679 PCT/US92/Ot539 replaced with 250 ~L of PBS containing BSA at a concentration of 1 mg per m~ and left at 4C for a further 1 hour. Prior to use in a phage binding assay the wells are washed rapidly 5 times with 250 ~L of PBS
containing 0.1~ I~reen.
5. B~nding ~ssays.
Bead A~Lay.
~ etween 10 and 20 ~L of the StrAv bead slurry (5 to 10 ~L bead volume) is washed 3 times with binding buffer (TBS containing BSA at a concentration of 1 mg per mL) just prior to the binding assay. 50 to 100 ~L of binding buffer containing control or peptide-display phage ( 1o8 to 10ll total plaque forming units - pfu's) is added to each microtube. Binding is allowed to proceed for 1 hour at room temperature using an end over end rotator. The beads are briefly centrifuged and the supernatant removed. The beads are washed a further 5 times with 1 m~ of TBS containing 0.1~ Tween, each wash consisting of a 5 min incubation and a brief centrifugation. Finally the bound phage are eluted from the StrAv beads by a 10 min incubation with pH 2 citrate buffer containing 1 mg per mL BSA which is subsequently neutralized with 260 ~L
of lM tris pH 8. The number of phage present in each step is determined as plaque forming units (pfu's) following appropriate dilutions and plating in a lawn of F' containing E. coli .
Plate ~say.
To each StrAv-coated well i9 added 100 ~L of binding buffer (PBS with 1 mg per mL BSA) containing a known quantity of phage (between 108 and 10ll pfu's). Incubation ~roceeds for 1 hr at room temperature followed by removal of the non-bound phage and 10 rapid wa9hes with PRS O . 1~
l~reen. The bound phage are eluted with 250 ~L of pH2 citrate buffer containing 1 mg per mL BSA and W092/1~679 PCT/US92/01~39 ~ ~ ~3 ;' c~ ~ ~

neutralization with 60 ~L of lM tris pH 8. The number of phage present in each step is determined as plaque forming units (pfu's) following appropriate dilutions and plating in a lawn of F' containing E. coli .
S EXAMPLE V
Effect of Dlth~othreltol (DTT) on display phage blnding to streptavldln-agarose.
Preliminary control experiments.
a. Use of HRP-conjugated biotin and streptavidin beads.
~inding capacity of StrAv agarose beads for HRP-conjugated biotin determined to be - 1 ~g (equivalent to - 150 pmol biotin) per 5 ~L beads (the amount used in these experiments).
b. Effect of DTT on HRP-conjugated biotin binding to StrA~ beads.
5 ~L of StrAv beads were incubated with 10 ng of HRP-biotin in binding buffer (T~S-BSA) in the presence of varying amounts of DTT (at least 99~ reduced). Following a 15 minute incubation at room temperature, the beads were washed two times in binding buffer and an HRP
substrate added. Color development was allowed to proceed and noted in a semi-quantitative manner. Table 827 shows that the binding of biotinylated horseradish peroxidase (HRP) is not greatly affected by concentrations of DTT below 20 mM. N.B. DTT
concentrations of 20 and 50 mM also inhibited the interaction of HRP and substrate in the absence of StrAv beads hence having a general negative effect in this system.

-- .

~, J
''I '~' ri~3 ~

~ffect of DTT on HPO6 display phage infectivity.

108 pfus of HPQ6 were added to binding buffer (T~S-BSA) in the pre~ence of different concentrations of DTT.
Incubated at room temperature for 1 hour then diluted and plated to determine titer as pfus. Table 828 show the effect of DTT on the infectivity of phage ~PQ6. Hence, either DTT has no effect on phage infectivities over this range of concentrations or the effects are reversed on dilution of the phage. From these control experiments it 10 i8 apparent that DTT can be used at concentrations below 10 mM in studies on the effect of reducing agents on peptide display phage binding to StrAv.

Table 829 shows the effect of DTT on the binding of phage HPQ6 and MXTN to StrAv beads. The most significant effect of DTT on HPQ6 binding to StrAv occurred between 0.1 and 1.0 mM DTT, a concentration at which no negative effects were observed in the preliminary control experiments. These results strongly indicate that, in the case of HPQ6 display phage, DTT has a marked effect on binding to StrAv and that the presence of a disulfide bridge within the displayed peptide is a requirement for good binding.
EXAMPLE ~I
RELEASE OF STREPTA~IDIN-BO~ND DISPLaY P~AGE
25BY ~ACTOR Xa CLEA~AGE
Phage HPQ6 contains a bovine F.X. recognition site (YIEGR/IV). In many instances, IEGR i8 sufficient recognition site for F.X" but we have extended the site in each direction to facilitate efficient cleavage. The effect of preincub~ting HPQ6 phage with F.X, on binding to StrAv beads is shown in Table 832. Thus while this concentration of F.X~ (2.5 units) had no measurable effect on the titer of the treated display phage it had a very marked effect on the ability of the treated display phage to bind to StrAv. This is consistent with the StrAv WO92/1~679 PCT/US92/01~39 recognition sequence being removed by the action of FX, recognizing and cleaving the YIEGR/IV sequence.
Table 833 shows the effect of FX~ treatment of HPQ6 following binding to StrAv. Is it possible to remove display phage bound to their target by the use of FX~ in place of p~ or chaotropic agent elution? HPQ6 display phage were allowed to bind to StrAv then incubated either in FX, buffer or the same buffer together with 2.5 units of FX, for 3 hrs. The amount eluted was compared to the total number of phage bound as judged by a pH2 elution.
Therefore, while the display phage are slowly removed in the buffer alone, the presence of FX, significantly increases this rate.
The removal of HPQ6 display phage from StrAv by FX, was also ~tudied as a function of the amount of enzyme added and the time of incubation, as shown in Table 834. N.B.
at greater concentrations of the enzyme (1.2 U for 1 hour or 2.5 U for 2 hours), a 109s in infectivity of the treated phage was noted as measured by pfus.

:- - . ~ . : .
~ . -,'' . ' , .' W092/15679 PCT/US92tOl~39 JC~
~Q~ 98 Table 10: Abundances obtained from various vgCodons A. Optimized fxS Codon, Restrained by [D]+~E] = [~]+[R]

T C A G _ l I.26 .18 .26 .30 f 2 1.22 .16 .40 .22 x 3 1.5 .0 .0 .5 S
Amino Amino acid Abundance acid Abun~nce A 4.80~ C 2.86 D 6.00~ E 6.00~
F 2.86~ G 6.60%
H 3.60~ I 2.86 K 5.20~ L 6.82 M 2.86~ . N 5.20 P 2.88~ Q 3.60 R 6.82~ S 7.02~ mf~a~
T 4.16~ V 6.60 W _ 2.86~ lfaa Y 5.20 stop 5.20~

21 0 a ~,i r) ~

99 :

[D] ~ [E] - [K] + [R] ~ .12 ratio - Abun(W)/Abun(S) = 0.4074 ` -(l/ratio)j ,(ratio)j sto,~-free 1 2.454 .4074 .9480 2 6.025 .1660 .8987 3 14.788 .0676 .8520 4 36.298 .0275 .8077 S 89.095 .0112 .7657 6 218.7 4.57 - 10-3 . 7258 7 536.8 1.86-103 .6881 .
, ' ' . : -W092/lS679 PCT/US92/OlS39 Table 10: Abundances obtained ~rom various vgCodon (continued) B. Unrestrained, optimized T C A G
1 1 .27 .19 .27 .27 2 1 .21 .15 .43 .21 3 1 .5 .0 .0 .5 Amino Amino acid Abundance acid Abundance A 4.05~ C 2.84%
D 5.81% E 5.81%
F 2.84% G 5.67 H 4.08% I 2.84 K 5.81~ L 6.83 M 2.84% N 5.81 P 2.85~ Q 4.08~
R 6.83% S 6.89~ mfaa T 4.05~ V 5.67~
W 2.84% lfaa Y 5.81%
8top 5.81%
~5 W092/t5679 PCT/US92/01~39 2 1 ~ 3 [D] + [E] = 0.1162 [K] + [R] - 0.1264 ratio = Abun(W)/Abun(S) = 0.41176 i (1 /rat io) j ( ratio ) J stop- f ree 1 2.4286 .41176 .9419 2 5.8981 .16955 .8872 3 14.3241 .06981 .8356 4 34.7875 .02875 .7871 84.4849 .011836 .74135 6 205.180 .004874 .69828 7 498.3 2.007 - 10-3 .6S77 -~ ~ . . .
' ~ , . . , ' .: ' .

W092tl5679 PCT/US92/01~39 Table 10: Abundances obtained from various vgCodon (continued) C. Optimized NNT

T C A G
1 1 .2071 .2929 .2071 .2929 2 1 .2929 .2071 .2929 .2071 3 1 1. Ø0 .0 Amino Amino acid Abundance acid Abundance A 6.06% C 4.29~ lfaa D 8.58% E none F 6.06% G 6.06%
H 8.58% I 6.06%
K none L 8.58%
2Q M none N 6.06%
P 6.06~ Q none R 6.06% S 8.58% mfaa T 4.29% lfaa V 8.58%
W none Y 6.06%
258top none - ' ..

, . ,' W092/1~679 PCT/US92/Ot~39 2t ~a~3 i (l/ratio)j (ratio)j stop-free 1 2.0 .5 1.
2 4.0 .25 1.
3 8.0 .125 1.
4 16.0 .0625 1.
5 32.0 .03125 1.
6 64.0 .015625 1.
7 128.0 .0078125 1.

.. . ..
. .
.
: , . ' ' ` ~ .
~: , W092/1~679 PCT/US9~tOlS39 ` 3 Table 10: Abundances obtained from various vgCodon (continued) D. Optimized NNG

T C A G
1 1 .23 .21 .23 .33 2 1- .215 .285 .2~5 .215 3 1 Ø0 .0 1.0 Amino Amino 15acid Abundance acid Abundance A 9.40~ C none D none E 9.40 F none G 7.10 H none I none K 6.60~ L 9.50~ mfa;a~
M 4.90~ N none P 6.00~ Q 6.00 R 9.50~ S 6.60 T 6.6 ~ V 7.10 W _ 4.90~ lfaa Y none stQp 6.60~

' ' ' ' w092/15679 PCT/US92/01539 i (l/ratio)j katioLj ~top-free 1 1.9388 .51579 0.934 2 3.7588 .26604 0.8723 ~ 7.2876 .13722 0.8148 4 14.1289 .07078 0.7610 27.3929 3.65 - l0-2 o .7108 6 53.109 1.88 lo-2 0.6639 7 102.96 9.72 ~ 1o-3 0 . 6200 , :
. .
:, , .

WO92/15679 PCT/US92/01539 .

~`;'''~

Table 10: Abundances obtained from optimum vgCodon (continued) ) E. Unoptimized NNS (NNK gives identical distribution) ~ T C A G
1 1 .25 .25 .2S.25 2 1 .25 .25 .25.25 3 1 .0 .5 .0 0.5 Amino Amino acid Abundance acid Abundance A 6.25~ C 3.125%
D 3.125~ E 3.125%
F 3.125~ G 6.25%
H 3.125% I 3.125%
X 3.125~ L 9.375%
M 3.125~ N 3.125%
P 6.25~ ' Q 3.125~
R 9.375~ S 9.375%
T 6.25~ V 6.25 W 3.125% Y 3.125 .

,. . . .

W092tl5679 PCT/US9~tO1539 ~1 Q~

stop 3.125~

i (l/ratio)J ~ratio)j sto~-free 1 3.0 .33333 .96875 2 9.0 .11111 .93~S
3 27.0 .03704 .90915 4 81.0 .01234S67 .8807 243.0 .0041152 .8532 6 729.0 1.37-103 .82655 7 2187.0 4.57 - 104 .8007 .
. .; . -- : -.. . .

W092/15679 PCT/US92/01i39 _ 39~3 Table 102b : Annotated Sequence of gene -after insertion of SalI linker nucleotide number 5'-(GGATCC TCTAGA GTC) GGC- 3 from pGEM polylinker tttac~ CTTTATGCTTCCGGCTCG tataat GTGTGG- 39 -35 lac W5 -10 aATTGTGAGCGcTcACAATT- 59 lacO-symm operator 20 g~g~ AGAGG CttaCT- 77 ~I Shine-Dalgarno ~eq.

¦fM ¦ X ¦ K ¦ S ¦ L ¦ V ¦ L ¦ X ¦ A ¦ S ¦
l 1 2 1 3 1 4 1 5 1 6 1 7 1 8 1 9 1 10l ¦ATG¦AAG¦AAA¦TCT¦~TG¦GTT¦CTT¦AAG¦GCT¦AGC¦ 107 .
- :- . . . .

. . ..... : ., . . . , - .

. . . . . . .

w092/l~679 PCT/US92/01~39 I Af1 II L~he I I

¦ V I A ¦ V I A I T I L I V I P I M I L I
S , I 11l 12l 13l 14l 15l 16l 17l 18l 19l 201 ¦GTT¦GCT¦GTC¦GCG¦ACC¦CTG¦GTA¦CCT¦ATG¦TTG¦- 137 ¦ NrU II I K~n I ¦

¦ S ¦ F ¦ A ¦ R ¦ P ¦ D ¦ F ¦ C ¦ L ¦ E ¦
21l 22l 231 241 251 26l 271 28l 291 301 ¦TCC¦TTC¦GCT¦CGT¦CCG¦GAT¦TTC¦TGT¦CTC¦GAG¦- 167 ~ ¦ACCIIII I A~a I ¦
M13 /BPTI Jnct ¦ Xho I P I P ¦ Y ¦ T I G I P ¦ C I K ¦ A ¦ R ¦
l 31I 321 331 341 351 361 371 381 391 401 lCC~ICC~IT~CIACTIaaalCCCITaCI~IaCalCaCI_ 197 I Pf1M ~ I 11 IL88H II¦
¦ A~a I ¦¦
L Dra II ¦
¦ P8S I

.
.. . .. .. . . . .
. . . . .. . . : - -. - : . . .. :

WO92/1~679 PCT/US92/01~39 Table 102b : Annotated Sequence of gene after insertion of ~l} linker (continued) R 1 Y 1 F ¦ Y ¦ N ¦ A ¦ K ¦ A ¦
421 431 441 451 461 471 481 491 50l ¦ATC¦ATC¦CGC¦TAT¦TTC¦TAC¦AAT¦GCT¦AAA¦GC 1- 226 G 1 ~ 1 C 1 Q ¦ T ¦ F ¦ V ¦ Y ¦ G ¦ G ¦

A¦GGC¦CTG¦TGC¦CAG¦ACC¦TTT¦GTA¦TAC¦GGT¦GGT¦- 257 1 Stu I¦ .
15¦ Xca I ¦

C 1 R 1 A 1 K 1 R ¦ N ¦ N ¦ F ¦ K ¦

20¦TGC¦CGT¦GCT¦AAG¦CGT¦AAC¦A~C¦TTT¦AAA¦ 284 I Esp I

¦ S ¦ A I E ¦ D ¦ C ¦ M 1 R 1 T 1 C 1 G
1 701 7l1 72~ 731 741 751 761 771 7~1 791 25¦TCG1GCC1GAA1GAT1TGC1ATG1CGT1ACC1TGC1GGT1 314 ¦XmaIII ¦ I Sph I¦

.. ~ , ... : . ........................ . .
.

~l ~a3~ ,~

BPTI/M13 boundary v I
¦ G ¦ A ¦ A ¦ E ¦ G ¦ D ¦ D ¦ P l A ¦ K l A ¦ A ¦
l 801 81l 82l 831 841 851 86l 871 88l 891 901 91I - :
¦GGC¦GCC¦GCT¦GAA¦GGT¦GAT¦GAT¦CCG¦GCC¦AAG¦GCG¦GCC¦- 350 ¦ Bbel ¦ t Sfi~
¦ Nar I ¦

l F ¦ N ¦ S ¦ L ¦ Q ¦ A ¦ S ¦ A ¦ T ¦
l 921 931 941 951 961 971 981 99l100l ¦TTC¦AAT¦TCT¦CTG¦CAA¦GCT¦TCT¦GCT¦ACC¦- 377 ¦Hind 31 ¦ E ¦ Y l I ¦ G ¦ Y l A l W l 1101l102l103l104l105l106l 1071 ¦GAGITATIATTIGGTITACIGCGITGGI- 3~8 l A ¦ M ¦ V ¦ V ¦ V ¦ I ¦ V ¦ G ¦ A ¦
1108l109l110l111l112l113l114l115l116l ¦GCC¦ATG¦GTG¦GTG¦GTT¦ATC¦GTT¦GGT¦GCT¦- 425 .¦ BstX I
.¦ Nco I¦

-.
, . ~ - . - ~ ' , . ,'' ' ' '', ' ' . . : '. '.

WO92/1~679 PCT/US92/01~39 9~ Q~ 3 Table 102b: Annotated Sequence after insertion of SalI linker (continued) ¦ T ¦ I ¦ G ¦ I ¦
117l118l119l120l ¦ACC¦ATC¦GGG¦ATC¦ 437 ¦ K ¦ L ¦ F ¦ K ¦ K ¦ F ¦ T ¦ S ¦ K ¦ A ¦

¦AAA¦CTG¦TTC¦AAG¦AAG¦TTT¦ACT¦TCG¦AAG¦GCG¦- 467 ¦A8U II¦

S I . I . I . I
l:L31l132l133l134l ¦TCT¦TAAITGAITAGI GGTTACC- 486 ~E II

terminator aTCGA GACctgca GGTCGACC ggcatgc-3' ¦SalI¦

.. : . ~ -: ~ . .,: :, , - . : , Wo 92/lS679 PCr/~'S92/01~39 21o~ ~

Note the f ollowing enzyme equivalences, XTna III = Eag I Acc III = ~M II
Dra II = EcoOl09 I ~a II = BstB I

, ,: ' ' ~ ' : : :

. : :

Wo 92/15679 PCI`/US92/OlS39 ~Irl N
~ _ _ 0 h ~I Cl ~1 ~
h H Wl ~ ~1 ~ Wl 1~ wl ~ Wl ~ m m ~ C ~ ~ ~ S t~ ~ ~ ~
~a v ~ m m m m m P~ m ~ u m m ~ ~ Q~

~ I V
.. ~
O --i V

m m m V S H ~1 ~41 Kl Kl H
H ~ H
Kl :~: h 4 D~ m m H H H
P3 ~ m H H H H H
--l Q~ ~ E R~ H H
~; o m ~

In o u~

. , . . ,. . : , . - , :

::

WO92/1~679 PCT/US92/01~9 lls~ ~- V9 3 3 ~

Table 107: In vitro transcription/translation analysis of vector-encoded signal::8PTI::mature VIII protein species _ _ 31 kd species' 14.5 kd ~peciesb_ No DNA (control) c pGEN-3Zf(-) +
pGEM-MB16 +
pGEM-MD20 + +
10 pGEM-MB26 + +
pGEM-M842 + +
pGEM-MB46 ND ND

Notes:
15 a.) pre-beta-lactamase, encoded by the amp ~bla) gene.
b.) pre-BPTI/VIII peptides encoded by the synthetic gene and derived constructs.
c.) - for absence of product; + for presence of product; ND for Not Determined.

.. , . , . : :
. ::..., - . .

.

WO9~/1;679 PCT/VS9'/Ot539 Table 108: Western analysis~ of ln v vo expressed signal::BPTI::mature VIII protein species A) expression in strain XLl-Blue _ siqnal 14.5 kd speclesb 12 kd cpecies'_ pGEM-3Zf(-) - d pGEM-MB16 VIII
pGEM-MB20 VIII ++
10 pGEM-MB26 VIII +++ +/-pGEM-MB42 phoA ++ +

B) expression in strain SEF' signal 14.5 kd speciesb 12 kd speciesC_ pGEM-MB42 phoA +/- +++

Notes:
a) Analysis using rabbit anti-BPTI polyclonal antibodies and horse-radish-peroxidase-conjugated goat anti-rabbit IgG
antibody.
b) pro-BPTI/VIII peptides encoded by the synthetic gene and derived constructs.
c) processed BPTI/VIII peptide encoded by the synthetic gene.
d) not present ......... -weakly present ...... +/-present ............. +
strong presence ..... ++
very strong presence +++

.

- .
.~

WO92tl~679 2 1 ~ .~ v ~3 ~ pcT/uss2/ol~3s Table 109: M13 gene III
1579 5'-GT GA~AAAATTA TTATTCGCAA TTCCTTTAGT

2051 GGTTCCGAAA TAGGCAGGGG GCATTAACTG m ATACGGG

2211 CTTTA~TGAG GATCCATTCG TTTGTGAATA TCAAGGCCAA

2411 ATGA~AAGAT GGCAAACGCT AATAAGGGGG CTATGACCGA

2571 TGGTGCTACT GGTGA m TG CTGGCTCTAA TTCCCAAATG

WO92/1~679 PCT~US92/01~39 ~> 118 Table 110: Introduction of ~EI into gene III
A) Wild-type III, portion encoding the signal peptide M K K L L F A I P

1579 5~-GTG AAA A~A TTA TTA TTC GCA ATT CCT TTA

/ Cleavage site V V P F Y S H S A E T V

1609 GTT GTT CCT TTC TAT TCT CAC TCC GCT GAA ACT GTT-3' B) II~, portion encoding the signal peptide with NarI site m k k 1 1 f a I p 1 :~-1579 5'-gtg aaa aaa tta tta ttc gca att cct tta / cleavage site v v p f y s G A a e t v 1609 gtt gtt cct ttc tat tct GGc Gcc gct gaa act gtt-3' , .. , . . . ~ . . ...................................... . .

. :
.. . . . . . , - . , WO92/15679 2 ~ ~9 ~3~ ~ PCT/US92/01539 Table 113 : Annotated Sequence of pGEM-MB42 comprising Ptac::~3S(G&AGGAAATAAA)::
DhoA-~ignal::mature-b~ti::mat~re-vIII-coat-protein 5'-GGATCC actccccatcccc ~HI

ctg TTGACA attaatcatcgGCTCG tataat GTGTGG--35 tac -10 aATTGTGAGCGcTcACA~TT-lacO-symm operator 20GAGCTCCATGGGAGA~AATAAA¦ATG~AAA¦CAA¦AGC¦ACGj-~SacI¦ c----- phoA signal peptide I A L ¦ L P L L F T P V T

ATC GCA CTC TTA CCG TTA CTG TTT ACC CCT GTG ACA
---------------- phoA signal conl :inu~ ~9 -- .___. ,___. , (There are no resldues 20-23.) ¦ ~ ¦ A ¦ R P ¦ D î F ¦ C ¦ ~ ¦ E î

phoA signal- ~A ¦ Acc: ~El ¦ A~ ra I
phoA/BPTI Jnct .¦ Xho I ¦
1~----- BPTI insert ---------¦ P ¦ P ¦ Y T G ¦ P ¦ C I K ¦ A I R
31 32 33 34 35l 36l 37l 38l 39l 40 lC~ ~ICCAl r~ClAc rlaaal ccl rocl~lccalcacl I p~M I l l lE3ss~ II¦
A~a I I
Dra II
Pss I

. . . : .

WO92/15679 PCT~S92/01539 ~.~Q V~ 120 ~ ' Table 113 : Annotated Sequence of Ptac::RBS( GGAGGAAATA~A) phoA-~ianal::mature-bpti::mature-VIII-coat-~rotein gene (continued) I I R Y F Y N A K A

ATC ATC CGC TAT TTC TAC AAT GCT AAA GC

G L C Q T F V Y G G

A GGC CTG TGC CAG ACC TTT GTA TAC GGT GGT
¦ Stu I¦ ACC I
Xca I
¦ C ¦ R A ¦ X ¦ R ¦ N ¦ N ¦ F X

TGC CGT GCT AAG CGT AAC AAC m AAA
Esp I
¦ S ¦ A ¦ E ¦ D ¦ C ¦ M ¦ R ¦ T I C I G I
1 70l 71l 72l 73l 74l 75l 76l 77l 78l 791 ¦TCG¦GCC¦GAA¦GAT¦TGC¦ATG¦CGT¦ACC¦TGC¦GGT¦-¦XmaIII¦ ¦_Sph I¦
-------------- BPTI insert-----------------BPTI/Ml3 boundary vl ¦ G ¦ A ¦ A ¦ E ¦ G ¦ D ¦ D ¦ P ¦ A I K I A I A I
1 801 8ll 82~ 831 841 85l 86l 871 881 891 gol 9ll GGCIGCCIGCT¦GAAIGGT¦GAT¦GAT¦CCG¦GCC¦AAG¦GCG¦GCC¦-1 ~3be I I ¦ Sfi I
I Na~ I ¦
-- BPTI--~¦c----- mature gene VIII coat protein ----¦ F ¦ N ¦ S ¦ L ¦ Q ¦ A I S I A I T I
1 921 931 941 951 961 971 981 991lO01 ¦TTC¦AAT¦TCT¦CTG¦CAA¦GCT¦TCT¦GCT¦ACC¦-lHind 8¦

¦ E ¦ Y ¦ I ¦ G ¦ Y I A I W ¦
llOll102110311041105110611071 ¦GAGITAT¦ATT¦GGT¦TAC¦GCG¦TGG¦-.. .. , . . ~
. .

. . ,. ~ .

WO92/lS679 21 Q a 3 ~ PCTIUS9~/01~39 Table 113 : Annotated Sequence of Ptac::RBS(GGAGGAAATAAA)::
~hoA-signal::mature-bpti::mature-vIXI-c,oat-~rotein gene (continued) ---¦ A ¦ M ¦ V ¦ V ¦ V ¦ I ¦ V ¦ G ¦ A ¦

GCC ATG GTG GTG GTT ATC GTT GGT GCT
~ Bst~ . I
I Nco I¦

¦117~118¦119¦120~
ACC ATC¦GGG ATC -X L F K K F T S X A

AAA CTG TTC AAG AAG TTT ACT TCG AAG GCG

11S31113211331134~
TCT TAA TGA TAG GGTTACC-B~tE II

AGTCTA AGCCCGC CTAATGA GCGGGCT T~ITTTTT-terminator ~g~ GACctgca GGTCGAC-3' ISalI¦

.. ~

. .

W O 92/lS679 ,? PC~r~US92/01539 Table 114: Neutralization of Phage Titer Using Agarose-immobilized Anhydro-Trypsin Percent Residual Titer As a Function of Time (hours) Phage Type Addition 1 2 4 MK-BPTI 5 ~l IS 99 104 105 2 ~l IAT 82 71 51 5 ~l IAT 57 40 27 10 ~1 IAT 40 30 24 MK 5 ~l IS106 96 98 2 ~l IAT 97 103 95 5 ~1 IAT 110 111 96 10 ~1 IAT 99 93 106 Begend:
IS ~ Immobilized streptavidin IAT = Immobilized anhydro-trypsin W~ ~/15679 2 ~ PCT/US92/01~39 Table 115: Affinity Selection of MX-~3PTI Phage on Immobilized Anhydro-Trypsin Percent of Total Phage 5 Phaqe TyDe Addition Recovered in Elution Buffer MX-BPTI5 ~l IS
2 ~l IAT 5 5 ~l IAT 20 10 ~l IAT 50 M~ 5 ~l IS c~l' 2 ~l IAT ccl 5 ~l IAT ccl 10 ~l IAT cc Legend:
IS ~ Immobilized streptavidin IAT ~ mobilized anhydro-tryp~in ' not detectable.

.. -, --, ~, :

.

' WO 92/lS679 PCr/VS9~/01;39 ~`3~
~ 124 Table 116: translation of Si~nal-III::E~s~::ma~ure-III : :
1 2 3 4 5 6 7 8 9 10 11 12 13 14 lS
fM K K L L F A I P L V V P F Y
GTG AAA AAA TTA TTA TTC GCA ATT CCT TTA GTT GTT CCT TTC TAT
¦c------- gene III signal peptide -------------------------S G A R P D F C L E P P Y T G
TCT GGC GCC cgt ccg gat ttc tgt ctc ga~ cca cca tac act ~g~
-----------~¦~----- BPTI insertion ------------------------P C X A R I I R Y F Y N A K A
ccc t~c aaa gcg cgc atç atc cgc tat ttc tac aat gct aaa gca G L C Q T F V Y G G C R A K R
qgc ctq tgç_cag acc ttt ~ta tac gqt ggt tgc cgt gct aag cqt N N F K S A E D C M R T C G G
aac aac ttt aaa tc~ gcc aaa ~at tgc ata cgt acc tgc ~gt qgc A G A A E T V E S C L A K P H
~cc GGC GCC GCT GAA ACT GTT GAA AGT TGT TTA GCA AAA CCC CAT
¦c------- mature gene III protein ---------------------91 92 93 94 g5 96 97 98 99 100 101 102 103 104 105T E N S F T N V W K D D K T L
ACA GAA AAT TCA TTT ACT AAC GTC TGG AAA GAC GAC AAA ACT TTA

: : - :. ................. . .

- :
: '' ' ~ ' : ~ : ' `:
.

WO92/156~9 2 ~ V' ~! :3 PCT/US92/01539 Table 116: tran~lation of Si~nal-III::bpti::mature-III
(continued) D R Y A N Y E G C h W N A T G
GAT CGT TAC GCT AAC TAT GAG GGT TGT CTG TGG AAT GCT ACA GGC

V V V C T G D E T Q C Y G T W
GTT GTA GTT TGT ACT GGT GAC GAA ACT CAG TGT TAC GGT ACA TGG

V P I G h A I P E N E G G G S
GTT CCT ~TT GGG CTT GCT ATC CCT GAA AAT GAG GGT GGT GGC TCT

E G G G S E G G G S E G G G T
GAG GGT GGC GGT TCT GAG GGT GGC GGT TCT GAG GGT GGC GGT ACT

K P P E Y G D T P I P G Y T Y
AAA CCT CCT GAG TAC GGT GAT ACA CCT ATT CCG GGC TAT ACT TAT

I N P L D G T Y P P G T E Q N
ATC AAC CCT CTC GAC GGC ACT TAT CCG CCT GGT ACT GAG CAA AAC

P A N P N P S h E E S Q P h N
CCC GCT AAT CCT AAT CCT TCT CTT GAG GAG TCT CAG CCT CTT AAT

T F M F Q N N R F R N R Q G A
ACT TTC ATG TTT CAG AAT AAT AGG TTC CGA AAT AGG CAG GGG GCA

WO92/1~679 PCT/US9'/01~39 ~ 126 Table 116: translation o~ Signal~ :b~ti::mature-III
(continued) L T V Y T G T V T Q G T D P V
TTA ACT GTT TAT ACG GGC ACT GTT ACT CAA GGC ACT GAC CCC GTT

K T Y Y Q Y T P V S S K A M Y
AAA ACT TAT TAC CAG TAC ACT CCT GTA TCA TCA A~A GCC ATG TAT

D A Y W N G K F R D C A F H S
GAC GCT TAC TGG AAC GGT AAA TTC AGA GAC TGC GCT TTC CAT TCT

G F N E D P F V C E Y Q G Q S
GGC TTT AAT GAG GAT CCA TTC GTT TGT GAA TAT CAA GGC CAA TCG

S D L P Q P P V N A G G G S G
TCT GAC CTG CCT CAA CCT CCT GTC AAT GCT C~GC GGC GGC TCT GGT

G G S G G G S E G G G S E G G
GGT GGT TCT GGT GGC GGC TCT GAG GGT GGT GGC TCT GAG GGT GGC

G S E G G G S E G G G S G G G
GGT TCT GAG GGT GGC GGC TCT GAG GGA GGC GGT TCC GGT GGT GGC

S G S G D F D Y E ~ M A N A N
TCT GGT TCC GGT GAT TTT GAT TAT GAA AAG ATG GCA AAC GCT AAT

,: ~ ' ' ' ~ - ' :~ . . , : . ., : ', ' ' WO92/l56~9 2 ~ Q a " i) t3 pcT/uss2/ols39 Table 116: translation of Si~nal-III::k~ mature-III
(continued) K G A M T E N A D E N A L Q S
AAG GGG GCT ATG ACC GAA AAT GCC GAT GAA AAC GCG CTA CAG TCT

D A X G ~ L D S V A T D Y G A
GAC GCT AAA GGC AAA CTT GAT TCT GTC GCT ACT GAT TAC GGT GCT

A I D G F I G D V S G L A N G
GCT ATC GAT GGT TTC ATT GGT GAC GTT TCC GGC CTT GCT AAT GGT

N G A T G D F A G S N S Q M A
AAT GGT GCT ACT GGT GAT TTT GCT GGC TCT AAT TCC CAA ATG GCT

Q V G D G D N S P L M N N F R
CAA GTC GGT GAC GGT GAT AAT TCA CCT TTA ATG AAT AAT TTC CGT

Q Y ~ P S ~ P Q S V E C R P F
CAA TAT TTA CCT TCC CTC CCT CAA TCG GTT GAA TGT CGC CCT TTT

V F S A G K P Y E F S I D C D
GTC TTT AGC GCT GGT AAA CCA TAT GA~ TTT TCT ATT GAT TGT GAC

,.

-WO92/15679 PCT/US92/01;39 ~Q 3 128 Table 116: translation of Signal~ :b~ti:: ature-III
(continued) 451. 452 453 454 455 456 457 458 459 460 461 462 463 464 465 K I N L F R G V F A F L L Y V
AAA ATA AAC TTA TTC CGT GGT GTC TTT GCG TTT CTT TTA TAT GTT
¦~----- uncharged anchor region -----A T F M Y V F S T F A N I L R
GCC ACC TTT ATG TAT GTA TTT TCT ACG TTT GCT AAC ATA CTG CGT
--------- uncharged anchor region continues ---------~¦

N K E S
AAT AAG GAG TCT TAA

Molecular weight of peptide = 58884 Charge on peptide = -20 [A+G+P] = 143 [C+F+H+I+~+M+V+W+Y] = 140 [D+E+K+R+N+Q+S+T~.] = 202 '' : , ' -- . , .

WO92/15679 ~ ~ 3 ~ ~J PCT/US92/0l~39 Table 116: translation o~ Siqnal-III :~E~i::mature-III
(continued) Second ~ase tc a . g t 15 21 15 8 t
12 5 10 6 c 4 0 0 a 0 30 4 g c 6 20 2 8 t 3 4 0 3 c 1 4 9 1 a 4 3 7 0 g a 5 19 21 1 t 4 11 1 c 2 4 16 1 a 8 2 4 2 g g 13 22 14 41 t 6 7 12 29 c 4 5 12 1 a 1 3 16 4 g AA # AA # AA # AA #

WO92/15679 PCT/US92/01~39 ~`v~ 130 Table 130: Sampling of a Library encoded by (NNK)6 A. Numbers of hexapeptides in each class total - 64,000,000 stop-free sequences.
can be one of [WMFYCIKDENHQ]
can be one of [PTAVG]
n can be one of [SLR] - -~a~ =2985984. ~aa~ =7464960.
n~ =4478976. ~ =7776000.
~n~ =9331200. nn~aa~ ~2799360.
~a~ =4320000. ~n~ =7776000.
~nn~a~ = 4665600. nQn~a~ = 933120.
=1350000. ~n~a =3240000.
~nQ~ =2916000. ~Qnn~ =1166400.
nnnn~ = 174960. ~ =225000.
~ n~ = 675000. ~nn~ =810000.
~Qnn~ = 486000. ~nnQQ~ =145800.
nnnnn~ = 17496. ~ = 15625.
n = 56250. ~ nn = 84375.
~nnn = 67500. ~Qnnn = 30375.
~nnnnn = 7290. nnnnnn = 729.
~nn~, for example, stands for the set of peptides ha~ing two amino acids from the ~ class, two from ~, and two from n arranged in any order. There are, for example, 729 = 36 sequences composed entirely of S, ~, and R.

. ' WO92/156~9 2 ~ 3 0 . PCT/US92/01~39 Table 130: Sampling of a Library encoded by (NN~) 6 (continued) B. Probability that any given stop-free DNA sequence will 5encode a hexapeptide from a stated class.
P ~ of class ~aaaa~...... 3.364E-03 (1.13E-07) ~a~...... 1.682E-02 (2.25E-07) na~a~...... 1.514E-02 (3.38E-07) ~aaaa...... 3.505E-02 (4.51E-07) ~naa~a . . . 6.308E-02 (6.76E-07) Qn~ . . . 2.839E-02 (l.OlE-06) ~...... 3.894E-02 (9.OlE-07) ~n~ . . .1 . 051E-01 (1.35E-06) ~Qn~aa...... 9.463E-02 (2.03E-06) QQQa~a...... 2.839E-02 (3.04E-06) aa...... 2.434E-02 (1.80E-06) ~Qaa...... 8.762E-02 (2.70E-06) ~QQaa...... 1.183E-01 (4.06E-06) ~nnQaa...... 7.097E-02 (6.08E-06) nQQQaa...... 1.597E-02 (9.13E-06) a...... 8.113E-03 (3.61E-06) ~ n~........ 3.651E-02 (5.41E-06) ~Qn~...... 6.571E-02 (8.11E-06) ~nQQ~...... 5.914E-02 (1.22E-05) QQa...... 2.661E-02 (1.83E-05) QQQQQ~...... 4.790E-03 (2.74E-05) ~ .......... 1.127E-03 (7.21E-06) ~ Q......... 6.084E-03 (1.08E-05) Qn ..... 1.369E-02 (1.62E-05) ~QQQ...... 1. 643E-02 (2.43E-05) ~QQQQ...... 1. lO9E-02 (3.65E-05) ~QQQQQ...... 3.992E-03 (5.48E-05) QQQQQn...... 5.988E-04 (8.21E-05) WO92/1~679 PCT/~S92/01~39 ~ 132 r~ , .
Table 130: Sampling of a Library encoded by (NNK) 6 (continued) C. Number of different stop-free amino-acid sequences in 5each class expected for ~arious library sizes Library size = l.OOOOE+06 total = 9.7446E~05 ~ sampled = 1.52 Cla~s Number ~Class Number ... 3362.6( .1) ~aa~..... 16803.4( .2) n~a~..... 15114.6( .3) ~a~..... 34967.8( .4) ~n~ ....... 62871.1( .7) Qna~ 28244.3( 1.0) ~ ......... 38765.7( .9) ~n~..... 104432.2( 1.3) ~QQ~a..... 93672.7( 2.0)QQQ~a..... 27960.3( 3.0) ... 24119.9( 1.8) ~Q~..... 86442.5( 2.7) ~nn~..... 115915.5( 4.0)~nnn~..... 68853.5( 5.9) nnnn~..... 15261.1( 8.7)~ ......... 7968.1~ 3.5) ~ Q~....... 35537.2( 5.3) ~Qn~..... 63117.5~ 7.8) ~nQn~..... 55684.4( ll.S) ~nnnQ~..... 24325.9( 16.7) QQQQn~..... 4190.6( 24.0) ~ ......... 1087.1( 7.0) n..... 5767.0( 10.3) ~ QQ....... 12637.2( 15.0) ~Qnn..... 14581.7( 21.6) ~nQQn..... 9290.2( 30.6) ~QnQQn..... 3073.9( 42.2) nnnnnn..... 408.4( 56.0) Library ~ize = 3.0000E+06 total = 2.7885E+06 ~ sampled ~ 4.36 a~ ........ 10076.4( .3) ~ ...50296.9( .7) n~ ........ 45190.9( 1.0) ~ ...104432.2( 1.3) ~na~..... 187345.5( 2.0) nn~...83880.9( 3.0) ~ aa.......115256.6( 2.7)~n~aa.....30,9107.9( 4.0) ~nn~..... 275413.9( 5.9)nnn~..... 81392.5( 8.7) ... 71074.5( s.3) ~n~.....252470.2( 7.8) ~Qn~..... 334106.2( 11.5) ~nnna~..... 194606.9( 16.7) nnnn~..... 41905.9( 24.0) ~ ......... 23067.8( 10.3) ~ na....... 101097.3( 15.0) ~nn~..... 174981.0( 21.6) ~nnn~..... 148643.7( 30.6) ~QQQn~..... 61478.9( 42.2) nnnnn~.... 9801.0( 56.0) ~ ......... 3039.6( 19.5) n.... 15587.7( 27.7) ~ nn....... 32516.8( 38.5) ~nnn.... 34975.6( 51.8) ~nnnn..... 20215.5( 66.6) ~nnnQQ.... 5879.9( 80.7) nnnnnn..... 667.0( 91.5) WO92/1567~ 2 ~ PCT/US92/01539 Table 130: Sampling of a Library encoded by (NNK) 6 (continued) Library size = l.OOOOE+07 total ~ 8.1204E+06 ~ sampled = 12.69 ...334S5.9( 1.1) ~a~ ....... 166342.4( 2.2) Qaaaaa..... 148871.1( 3.3) ~aaaa..... 342685.7( 4.4) ~Q~aaa..... 609987.6( 6.5) nQaaaa..... 269958.3( 9.6) ~a~a..... 372371.8( 8.6) ~n~a..... 983416.4( 12.6) ~nnaaa..... 856471.6( 18.4) nnn~aa..... 244761.5( 26.2) aa..... 222702.0( 16.5) ~Qaa..... 767692.5( 23.7) ~nn~a..... 972324.6( 33.3) ~nnnaa..... 531651.3( 45.6) nnnna~..... 104722.3( 59.9) ~ a........ 68111.0( 30.3) na..... 281976.3( 41.8) ~nna..... 450120.2( 55.6) ~nnna..... 342072.1( 70.4) ~nnnna..... 122302.6( 83.9) nnnnn~..... 16364.0( 93.5) ~ ......... 8028.0( 51.4) ~ n. . .37179.9( 66.1) ~ nn. . .67719.5( 80.3) ~nnn..... 61580.0( 91.2) ~nnnn..... 29586.1( 97.4) ~nnnnn. . .7259.5( 99.6) nnnnnn. . .728.8(100.0) Library size = 3.0000E+07 total = 1.8633E+07 ~ sampled 8 29.11 aaaaaa......... 99247.4( 3.3) ~aa~a.... 487990.0( 6.5) n~ a.......... 431933.3~ 9.6) ~aaa~.... 983416.5( 12.6) ~na~a~...... 1712943.0( 18.4) ~naaaa.... 7342B4.6( 26.2) ~aaa...... 1023590.0( 23.7) ~naa~.... 2592866.0( 33.3) ~nnaaa...... 2126605.0( 45.6) nnnaaa.... 558519.0( 59.9) a....... 563952.6( 41.8) ~naa.... 1800481.0( 55.6) ... 2052433.0( 70.4) ~nnnaa.... 978420.5( 83.9) nnnna~.... 163640.3( 93.5) ~ a....... 148719.7( 66.1) ~ Qa...... 541755.7( 80.3) ~nna.... 738960.1( 91.2) ~Qnna.... 473377.0( 97.4) ~nnnQa.... 145189.7( 99.6) nnnnna.... 17491.3(100.0) ~ ........ 13829.1( 88.5) n.... 54058.1( 96.1) ~ nn...... 83726.0( 99.2) ~nnn.... 67454.5( 99.9) ~nnnn.... 30374.5(100.0) 40 ~nnnnn.... 7290.0(100.0) nQnnnQ.... 729.0(100.0) '. - ~ - - -, W092/1~679 PCT~US9~tO1539 ~ 3~-' Table 130: Sampling of a Library encoded by (NNK) 6 (continued) hibrary size = 7.6000E+07 total - 3.2125E+07 ~ sa~pled 8 50.19 a~ .......... 245057.8( 8.2) ~a~..... 1175010.0( 15.7) n~ ......... 1014733.0( 22.7) ~ ......... 2255280.0( 29.0) o ~n~ ........ 3749112.0( 40.2) nn~ ....... 1504128.0( 53.7) ... 2142478.0( 49.6) ~Q~..... g993247.0( 64.2) ~nna~...... 3666785.0( 78.6) nnn~..... 840691.9( 90.1) ... 1007002.0( 74.6) ~n~..... 2825063.0( 87.2) ~nn~...... 2782358.0( 95.4) ~Qnn~..... 1154956.0( 99.0) lS nnnnaa..... 174790.0( 99.9) ~ ......... 210475.6( 93.5) n~..... 663929.3( 98.4) ~Qn~..... 808298.6( 99.8) ~nQn~..... 485953.2(100.0) ~QnnQ~..... 145799.9(100.0) QQnQQ~..... 17496.0(100.0) ~ ......... 15559.9( 99.6) ~ Q. . .56234.9(100.0) ~ QQ. . .84374.6(100.0) ~QnQ..... 67500.0(100.0) ~nnQn..... 30375.0(100.0) ~nQQQn..... 7290.0(100.0) nQnQnn..... 729.0(100.0) hibrary size ~ l.OOOOE+08 total = 3.6537E+07 % sampled = 57.09 ~a~a....... 318185.1( 10.7) ~aaa~..... 1506161.0( 20.2) Qaaaaa....... 1284677.0( 28.7) ~aaaa..... 2821285.0( 36.3) ~Qa~a....... 4585163.0( 49.1) nQ~aaa..... 1783332.0( 63.7) ~ ........... 2566085.0( 59.4) ~n~..... 5764391.0( 74.1) ~nQ~....... 4051713.0( 86.8) QQn~..... 888584.3( 95.2) ... 1127473.0( 83.5) ~n~..... 3023170.0( 93.3) ~QQ~...... .2865517.0( 98.3) ~nnn~..... 1163743.0( 99.8) QnQn~..... 174941.0(100.0) ~ ......... 218886.6( 97.3) ~ n~....... 671976.9( 99.6) ~Qn~..... 809757.3(100.0) ~nQQ~..... 485997.5(100.0) ~QQQn~..... 145800.0(100.0) nnQnQ~..... 17496.0(100.0) ~ ......... 15613.5( 99.9) n..... 56248.9(100.0) ~ nn....... 84375.0(100.0) ~nnn..... 67500.0(100.0) ~nnnn..... 30375.0(100.0) ~nQnnn..... 7290.0(100.0) nnnnnn..... 729.0(100.0) . : . , , " '' ', W092/15679 ~t ~ ~ 3 ~ ~ PCT/USg2/01539 Table 130: Sampling of a Library encoded by (NNK) 6 (continued) Library size ~ 3.0000E+08 total ~ 5.2634E+07 ~ sampled 8 82.24 ... 856451.3( 28.7) ~ ...3668130.0~ 49.1) n~a~.~. 2854291.0( 63.7) ~ ...5764391.0( 74.1) o ~n~aa~...... 8103426.0( 86.8) nn~a.....2665753.0( 95.2) a~...... 4030893.0( 93.3) ~na~.....7641378.0( 98.3~
~nna~a...... 46S4972.0( 99.8) nnna~.....933018.6(100.0) ... 1343954.0( 99.6) ~n~..... 3239029.0(100.0) ~nQ~...... 2915985.0(100.0) ~nnn~..... 1166400.0~100.0) nnnn~..... 174960.0(100.0) ~ ......... 224995.5(100.0) n~..... 674999.9tlOO.O) ~nn~..... 810000.0(100.0) ~nnn~..... 486000.0(100.0) ~nnnn~..... 145800.0(100.0) nnnnn~..... 17496.0(100.0) ~ ......... 15625.0(100.0) ~ n........ 56250.0(100.0) ~ nQ....... 84375.0~100.0) ~nnn..... 67500.0(100.0) ~nnnn..... 30375.0(100.0) ~nnnnn..... 7290.0(100.0) nnnnnn..... 729.0(100.0) ~ibrary size ~ l.OOOOE+O9 total = 6.1999E+07 ~ sampled e 96.87 ... 2018278.0( 67.6) ~ ... 6680917.0( 89.S) n~ ......... 4326519.0~ 96.6) ~ ... 7690221.0( 98.9) ~n~a~...... 9320389.0( 99.9) nn~ ... 2799250.0(100.0) ... 4319475.0(100.0) ~n~..... 777S990.0(100.0) Q~...... 4665600.0(100.0) nnn~..... 933120.0(100.0) ... 1350000.0(100.0) ~n~..... 3240000.0(100.0) ~nn~...... 2916000.0(100.0) ~nnn~..... 1166400.0(100.0) nnnna~..... 174960.0(100.0) ~ ......... 225000.0(100.0) n~..... 675000.0(100.0) ~nn~..... 810000.0(100.0) ~nnn~..... 486000.0(100.0) ~nnnn~..... 145800.0(100.0) nnnnn~..... 17496.0(100.0) ~ ......... 15625.0(100.0) ~ n........ 56250.0(100.0) ~ nQ....... 84375.0(100.0) ~nQn..... 67500.0(100.0) ~nnnn..... 30375.0(100.0) ~QnnQn..... 7290.0(100.0) QQnnnn..... 729.0(100.0) ... .. ,, ~
. ~ :.. . . -.

: -~ :: : . ' : ' .

W O 9'/1~679 PC~r/US9'/01539 ~ ~Q 136 c~ ~,Q ~
Table 130: Sampling of a Li~rary encoded by (NNK) 6 (continued) Library size = 3.0000E+09 total = 6.3a90E+07 ~ sampled - 99.83 aa~a~a...... 2884346.0( 96.6) ~aaa~...... 7456311.0( 99.9) Qaaaaa...... 4478800.0(100.0) ~a~...... 7775990.0(100.0) ~Qaaaa...... 9331200.0(100.0) QQaolao~.... 2799360.0(100.0) ... 4320000.0(100.0) ~Q~...... 7776000.0(100.0) ~Qn~aa...... 4665600.0(100.0) QQQaaa...... 933120.0(100.0) a...... 1350000.0(100.0) ~Q~...... 3240000.0(100.0) ~QQaa...... 2916000.0(100.0) ~QQQaa...... 1166400.0(100.0) nQnQa~..... 174960.0(100.0) ~ a......... 225000.0(100.0) n~. . . 675000.0(100.0) ~nn~. . . 810000.0(100.0) ~nnn~. . . 486000.0(100.0) ~nQQna...... 145800.0(100.0) QQQnn~...... 17496.0(100.0) ~ .......... 15625.0(100.0) ~ n......... 56250.0(100.0) ~ QQ........ 84375.0(100.0) ~nQn...... 67500.0(100.0) ~QQQn...... 30375.0(100.0) ~nnnQQ...... 7290.0(100.0) nnnnnn . . . 729.0(100.0) W092/l5679 ~ PCT/US92/01539 Table 130, continued D. Formulae for tabulated quantities.
S L~ize is the number of independent transformants.
31**6 is 31 to sixth power; 6*3 means 6 times 3.
A - ~size/(31**6) can be one of [WMFYCIKDENHQ.]
can be one of [PTAVG]
o n ca~ be one of [S~R]
F0 - (12)**6 F1 ~ (12)**5 F2 ~ (12)**4 F3 - (12)**3 F4 ~ (12)**2 F5 = (12) 15 aaaaaa = F0 * (1-exp(-A)) ~aaaaa - 6 * 5 * F1 * (1-exp(-2*A)) naa~aa = 6 * 3 * F1 * (1-exp(-3*A)) ~aaaa = (15) * 5**2 * F2 * (l-exp(-4*A)) ~naaaa = (6*5)*5*3 *F2 * (1-exp(-6*A)) nnaaa~ = (15) * 3**2 * F2 * (1-exp(-9*A)) = (20)*(5**3) * F3 * (1-exp(-8*A)) ~n~aa ~ (60)*(5*5*3)*F3* (1-exp(-12~A)) ~nnaaa ~ (60)*(5*3*3)*F3*(1-exp(-18*A)) nnnaaa = (20)*(3)**3*F3*(1-exp(-27*A)) ~ aa = (15)*(5)**4*F4*(1-exp(-16*A)) ~naa ~ (60)*(5)**3*3*F4*(1-exp(-24*A)) aa = (90)*(5*5*3*3)*F4*(1-exp(-36*A)) ~nnnaa ~ (60)*(5*3*3*3)*F4*(1-exp(-54*A)) nnnnaa = (15)*(3)**4 * F4 *~1-exp(-81*A)) ~ a ~ (6)*(5)**5 * F5 * (l-exp(-32*A)) na = 30*5*5*5*5*3*F5*(1-exp(-48*A)) ~nna = 60*5*5*5*3*3*F5*(1-exp(-72*A)) ~nnn~ = 60*5*5*3*3*3*F5*(1-exp(-108*A)) ~nnnna = 30*5*3*3*3*3*F5*(1-exp(-162*A)) nnQnna = 6*3*3*3*3*3*F5*(1-exp(-243*A)) = 5**6 * (1-exp(-64*A)) Q ~ 6*3*5**5*(1-exp(-96*A)) nn ~ 15*3*3*5**4*(1-exp(-144*A)) ~nnn = 20*3**3*5**3*(1-exp(-216*A)) ~nnnn ~ 15*3**4*5**2*(1-exp(-324*A)) ~nnnnn - 6*3**5*5*(1-exp(-486*A)) nnnnnn - 3**6*(1-exp(-729*A)) total ~ aaaaaa + ~aaaaa + naaaaa ~ ~aaaa + ~naaaa +
nna~ + ~aaa + ~na~ + ~nnaa~ + nnnaa~ +
~ aa + ~naa + ~nn~ + ~nnna~ + nQnn~ +
a + ~ Qa + ~QQa + ~nnna + ~Qnnna +
nnQnn~ + ~ + ~ n + ~ nn + ~nQn +
~nnnn + ~nQQnn + nnnQQn Table 131: Sampling of a Library Encoded by (NNT)~(NNG)2 X can be F,S,Y,C,~,P,H,R,I,T,N,V,A,D,G
r can be L',R2,S,W,P,Q,M,T,K,V,A,E,G
Library comprises 8.55-l06amino-acid sequences; 1.47 107DNA
sequences.
Total number of possible aa sequences= 8,555,625 x LVPTARGFYCHIND
S S
~ VPTAGWQMKES
n LR

The first, second, fifth, and sixth positions can hold x or S; the third and fourth position can hold a or n. I
have lumped sequences by the number of xs, S9, Og, and Qs.
For example xx~nSS stands for:
[xxanSS, xSanxS, xSanSx, ssonxx, Sx~QxS, sxanSx, xxQ6SS, xSn~xS, xSn~Sx, ssnaxx, sxnoxs~ sxnosx]
The following table shows the likelihood that any particular DNA sequ~nce will fall into one of the defined classes .
~ibrary size - 1.0 Sampling = .00001~
total........... 1.OOOOElO0 ~sampled........ 1.1688E-07 xxa~xx.......... 3.1524E-01 xx~nxx~ 2.2926E-01 xxnnxx~ 4.1684E-02 xx~xS.......... 1.8013E-01 xxanxs.......... 1. 3101E-01 xxnnxs.......... 2.3819E-02 xx60SS.......... 3.8600E-02 xxonss.......... 2.8073E-02 xxnnss.......... 5.1042E-03 xS~SS.......... 3.6762E-03 xsanss.......... 2.6736E-03 xsnnss.......... 4.8611E-04 SSOOSS.......... 1.3129E-04 ssonss.......... 9.5486E-05 ssnnss.......... 1. 7361E-05 , ~ ~, ~ , : ' ' ' ' ' ' : . ' . .

WO92~l;679 ~1 ~ a~ PCT/VS92/01539 Table 131: Sampling o~ a Library Encoded by (NNT)4(NNG)2 (continued) The following sections show how many ~equences of each class are expected for libraries of different sizes.

Library size ~ l.OOOOE+05 total........ 9.9137E+04 fraction sampled - 1.1587E-Q2 Type Number ~ Type Number xxOOxx....... 31416.9( .7) xx~nxx........ 22771.4( 1.3) xxnnxx....... 4112.4(2.7) xx90xS........ 17891.8( 1.3) xxsnxs....... 12924.6(2.7) xxnnxs........ 2318.5( 5.3) xxOOSS....... 3808.1(2.7) xxsnss........ 2732.5( 5.3) xxnnss....... 483.7( 10.3) xsaoss........ 357.8( 5.3) xsonss....... 253.4( 10.3) xsnnss........ 43.7( 19.5) SSOOSS....... 12.4( 10.3) ssonss........ 8.6( 19.5) ssnnss....... 1.4( 35.2) ~ibrary size - l.OOOOE+06 total........ 9.2064E+05 fraction sampled = 1.0761E-01 xxOOxx.......304783.9( 6.6) xxonxx........ 214394.0( 12.7) xxnnxx....... 36508.6( 23.8) xxOOxS....... 168452.5( 12.7) xx9nxS.......114741.4( 23.8) xxnnxS....... 18383.8( 41.9) xx~aSS....... 33807.7( 23.8) xx~nSS....... 21666.6( 41.9) xxnnss....... 3114.6( 66.2) xSaOSS....... 2837.3( 41.9) xsonss....... 1631.5( 66.2) xsnnss....... 198.4( 88.6) SSOOSS....... 80.1( 66.2) ssanss....... 39.0( 88.6) ssnnss....... 3.9( 98.7) Library size . 3.0000E+06 total........ 2.3880E+06 fraction sampled . 2,7912E-01 xx~Oxx.......855709.5( 18.4) xxonxx....... 565051.6( 33.4) xxnnxx........85564.7( 55.7) xxOOxS....... 443969.1( 33.4) xxonxs.......268917.8( 55.7) xxnnxs....... 35281.3( 80.4) xx~OSS........79234.7( 55.7) xxOnSS....... 41581.5( 80.4) xxnnss.........4522.6( 96.1) xSOOSS....... 5445.2( 80.4) xsonss.........2369.0( 96.1) xsnnss....... 223.7( 99.9) SSOOSS.........116.3( 96.1) ssonss....... 43.9( 99.9) SSQQSS......................... 4.0(100.0) . .
- , ~ ~. . . . . - - , . , ~ , ~ , . .

.
. .

W092/15679 ~ PCT/US92/01~39 J ~i`3 Table 131: Sampling of a Library Encoded by (NNT)~NNG) 2 (continued) 5Library size = 8.5556E+06 total........ 4.9303E+06 fraction sampled = 5.7626E-Ol xx~xx....... 2046301.0( 44.0) xxonxx...... 1160645.0( 68.7) xxnnxx....... 138575.9( 90.2) xx~xS....... 911935.6( 68.7) x~nxs....... 435524.3( 90.2) xxnQxs....... 434aO.7( 99.0) xxaoSs....... 128324.1( 90.2) xxonss....... 51245.1( 99.0) xxnnss....... 4703.6tlOO.O) xSO~SS....... 6710.7( 99.0) xsonss....... 2463.8(100.0) xsnnss....... 224.0(100.0)SS03SS....... 121.0(100.0) ssonss....... 44.0(100.0) ssnnss....... 4.0(100.0) Library size = l.OOOOE+07 total........ 5.3667E+06 fraction sampled = 6.2727E-01 xx~xx....... 2289093.0( 49.2) xx~nxx...... 1254877.0( 74.2) xxnnxx....... 143467.0( 93.4) xx~OxS....... 985974.9( 74.2) xxOnxS....... 450896.3( 93.4) xxnQxS....... 43710.7( 99.6) xxOOSS....... 132853.4( 93.4) xxonSS....... 51516.1( 99.6) xxnnss....... 4703.9(100.0) xS~SS....... 6746.2( 99.6) xsonss....... 2464.0(100.0) xSQQSS....... 224.0(100.0) SS~SS....... 121.0(100.0) SS0QSS....... 44.0(100.0)ssnnss....... 4.0(100.0) Library size - 3.0000E+07 total........ 7.8961E+06 fraction sampled = 9.2291E-01 xx~Oxx....... 4040589.0( 86.9) xxonxx...... 1661409.0( 98.3) xxnnxx...... 153619.1(100.0) xxOOxS....... 1305393.0( 98.3) xxonxs......482802.9(100.0) xxnnxs....... 43904.0(100.0) xxOOSS......142254.4(100.0) xx~nss....... 51744.0(100.0) xxnnss...... 4704.0(100.0) xSO~SS....... 6776.0(100.0) xS~QSS...... 2464.0(100.0) xsnnss....... 224.0(100.0) SS~SS...... 121.0(100.0) SS~QSS....... 44.0(100.0) ssnnss...... 4.0(100.0) , . . . . .

. ~
' ~ ' ' ~ ' ' , - . .

WO92/1~679 2 ~ cl ~!? `, PCT/US92/01~39 Table 131: Sampling of a Library Encoded by (NNT) 4 (NNG)2 (continued) SLibrary size ~ 5.0000E+07 total....... 8.3956E+06 fraction sampled - 9.8130E-01 xx~Oxx...... 4491779.0( 96.6) xx~nxx...... 1688387.0( 99.9) xxnnxx...... 153663.8(100.0) xx90xS....... 1326590.0( 99.9) xxanxs......482943.4(100.0) xxnnxs....... 43904.0(100.0) xxasss......142295.8(100.0) xxOnSS....... 51744.0(100.0) xxnnss...... 4704.0(100.0) xSOOSS....... 6776.0(100.0) xs3nss~ . 2464.0(100.0) xSnnSS....... 224.0(100.0) ssaoss...... 121.0(100.0) ssonss....... 44.0(100.0) ssnnss...... 4.0(100.0) Library size = l.OOOOB+08 total....... 8.5503E+06 fraction sampled = 9.9938E-01 xx3~xx...... 4643063.0( 99.9) xxOnxx...... 1690302.0(100.0) xxnnxx...... 153664.0(100.0) xx~0xS....... 1328094.0(100.0) xx3nxS......482944.0(100.0) xxnnxS....... 43904.0(100.0) xx~OSS .....142296.0(100.0) xxOnSS....... 51744.0(100.0) xxnnss:::... 4704.0(100.0) xS~OSS....... 6776.0(100.0) xS~QSS...... 2464.0(100.0) xsnnss....... 224.0(100.0) ss0ass...... 121.0(100.0) ss6nss....... 44.0(100.0) ssnnss........ 4.0(100.0) .... - ~ -.
.

WO 92/1~679 PCr/US9~/Ot;39 Table 132: Relative efficiencies of various simple variegation codons Nu~er of codons #DNA/#AA #DNA/#AA #DNA/#AA
[#DNA] [~DNA][#DNA]
vaCodon (#AA) (#AA) (#AA) 10 NN~ 8.95 13.86 21.49 assuming [2.86-107] [8.87- lot] [2.75-10~]
stops vanish (3.2-106)(6.4-107) (1.28-109) NNT 1.38 1.47 1.57 [1.05-106] [1.68-10~] [2.68-10~]
(7.59-105) (1.14-107) (1.71-10t) NNG 2.04 2.36 2.72 , assuming [7.5910l] [1.14 106] [1 . 71 lOt]
20 stops vanish (3.7-105)(4.83-106) (6.27-107) WO g'/1~679 2 1 0 ~ ~ ~J .j PCT/US9'/01~39 Table 140. Effect of antl 9PTI IgG on phaye titer.
Phage Input +Anti-BPTI +Anti-BPTI Eluted Phage Str~in +Protein A(a) M13MP18 100 (b) 98 92 7-104 5 BPTI.3 100 26 21 6 M13MB48 (c) 100 90 36 0.8 M13MB48 (d) 100 60 40 2.6 (a) Protein A-agarose beads.
(b) Percentage of input phage measured as plaque forming units (c) Batch number 3 (d) Batch number 4 Table 141. E fect of antl-BPTI or prote~n A on phage tlter.
No +Anti- +Anti-Strain Input Addition 8PTI +Protein A BPTI
(a~+Protein A
M13MP18 lOO(b) 107 105 72 65 20 M13MB48(b)100 _ 92 7.103 58 ~10 (a) Protein A-agarose beads (b) Percentage of input phage measured as plaque forming units (c) Batch number 5 .
.: , . ' '. `: '.
: .

Wo92/15679 PCT/VS92/OtS39 ~ 144 Tabl~ 142 Effect of anti-B~TI and non-lmmune serum on phage tlter +Anti- +Anti- +NRS
5 Strain InputBPTI +NRS BPTI IProtein (a) +Protein A A
(b) M13MP18 lOO(c)65 104 71 88 M13MB48(d) 10030 125 13 121 M13MB48(e) 100 2 105 0.7 110 (a) Purified IgG from normal rabbit serum.
(b) Protein A-agarose beads.
(c) Percentage of input phage measured as plaque forming units (d) Batch number 4 (e) Batch number 5 Table 143. Lo~ ln tlter of dlRplay phage with anhydrotryp~ln.
AnhydrotrypsinStrepta~idin Strain Beads Beads Post Post Staxt _ Incubation ~tart Incubation M13MP18 100 (a) 121 ND ND

5~A Pool 100 44 100 93 (a) Plaque forming units expressed as a percentage of input.

..
, ..: ~ . .
.

WO92t15679 PCT/~S92/01539 21 ~ a ~

Table 144. Binding of Display Phage to Anhydrotryp~in.

Experiment 1.
StrainEluted Phage (a)Relative to M13MP180.2 (a) 1.0 BPTI-IIIMK 7.9 39.5 M13M84811.2 56.0 Experiment 2.
StrainEluted Phage ~a)Relative to M13mpl8 M13mpl~ 0.3 1.0 BPTI-IIIMK 12.0 40.0 M13MB5617.0 56.7 (a) Plaque forming units acid eluted from beads, expressed as a percentage of the input.

Table 145. Blndlng of Display Phage to Anhydrotrypsin or Trypsln.
StrainAnhydrotrv~sin BeadsTrypsin Beads Eluted Eluted Phage Relative Phage Relative (a)8indina (b) Bindinq MI3MP18 ¦ 0.1 1¦ 2.3x104 1.0 BPTI-IIIMK ¦ 9.1 91 ¦ 1.175x103 M13.3X7 ¦ 25.0 250 ¦ 1.46x103 M13.3X11 ¦ 9.2 92 ¦ 0.271.2x103 (a) Plaque forming unit~ eluted from beads, expressed as a percentage of the input.
(b) Relative to the non-display phage, M13MP18.

.

: , ,. . ~. ' ...... . .
.. . ~. . . ~ : ' ' , ~ ' '' ~- " ' ' WO9~/15679 ~ PCT/US92/01539 ~`.3~' ~p 146 Table 146. B~ndlng of Dlsplay Phage to Trypsln or H~An Neutrophll Elastase.
StrainTrypsin Beads ¦ HNE Beads 5 Eluted Phage Relative ¦ Eluted Relative (a) Binding(b)l Phage Binding M13MP18 ¦ 5xlO~ 1 ¦ 3x104 1.0 BPTI-IIIMK¦ 1.0 2000 ¦ 5x103 i6.7 10 M13MB48 ¦ 0 13 260 ¦ 9x10~3 30.0 .

M13.3X7 ¦ 1.15 2300 ¦ lx10~3 3.3 M13.3X11¦ 0.8 1600 ¦ 2x103 6.7 BPTI3.CL¦ lX10 3 2 ¦ 4.1 1.4xlO~
(c) (a) Plaque forming units acid eluted from the beads, expressed as a percentage of input.
(b) Relative to the non-display phage, M13MP18.
(c) BPTI-IIIMK (K15L MGNG) WO92/1~679 PCT/US92/01539 2 ~

Table 820: Streptavldln-Blndlng P~age Putative Streptavidin Name Bindinq Peptide Se~.
DEV(F) A E - P C H P O Y R L C Q R P L K Q P P P P P P A E...
Dev(E) A E - L C H P O F P R C N L F R K V P P P P P P A E...
HPQ6 A E G P C H P O F P R C Y I E G R I V - - - - - - E...

- - - - C - - - - - - C - - - - - - - - - - - - - E

,, , . - - ~ : , . -., . . . . ........................ - .
.: . . .

WO 92/1~679 r~ PCI`/US92/Ot~39 .~3 Table 827z E~fect of DTT on blotlnylated ~RP -:
blndlng to ~treptavldln agarose.
Conc. DTT ~iotin-HRP Color _ _ (mM) Develo~ment O ++++
2 ++++
+++
+

Table 828: Effect of DTT on ~PQ6 dlsplay phage ~nfectlvlty.
DTT (mM) Relative Infectivity 0 1.00 2 0.95 1.00 1 . 08 .

WO92/156792 ~ f~ ~ PCT/US92/01~39 1,23 Table 829: Effect of the pre~ence of -:
DTT on the blndlng of dlsplay phage to ~troptavldln agaroso beads. ::.
Input~: MXTN 4.2 x 10ll, HPQ6 3.3 x loll.
NameConcn DTT Fraction Bound Relative ~M) ~3indinq MKTN O 4.8 x 10~ 1.00 2,000 5.4 x 10~ 1.10 10,000 5.2 x 10~ 1.10 HPQ6 0 1.6 x 104 1.00 1 1.6 x 104 1.00 1.5 x 10~ 0.90 100 9 . 7 X 10'5 0 . 60 1000 1.6 x 10'5 0 . 10 2, 000 1 . 0 X 10'5 0 . 06 5.000 8.8 x 104 o.o5 ::

WO92/15679 ~ PCT/US9'/01539 Table 832s Efect of prelncubatlng ~PQ6 dlsplay ph~ge with Factor X. on b~ndlng to streptavldln beads.
Titer after Relative Relative Factor X, Treatment Titer Fraction Bound 9indinq - 3.3 x 10ll 1 1.4 x 104 1 .
+ 3.3 x 10ll 1 1.2 x 105 8.5 x lo-2 Table 833s FX, treatment o ~PQ6 dlsplay phage followlng blndlng to streptavld~n.

Total Fraction~ Removed Factor X, Fraction Eluted by Bound by Treatment Treatment - 7.6 x 10'3 1.6 x 1o-3 14 + 6.4 x 103 2.6 x 1o-3 40 Table 834s Remo~al of ~PQ6 dlsplay phage from streptavldln by FX, ~mount of FX, Time % Removed by ~dded (unit~) ~hrs) Treatment.

2.5 1 21 6.3 1 22 12.5 1 35 2.5 2 53 6.3 2 54 12.5 2 52 `

WO92/15679 21 ~ ~ 3 ~9 `3 PCT/US92/01;39 lS1 CITATIONSSHM89: Ashman, Matthew~, and Frank (1989) Protein Engineering 2(5): 387-91.ANN81: Banner, DW, C Nave, and DA Marvin, Nature (1981), 289:814-816.ECK89c: Becker, S, E Atherton, and RD Gordon, Eur J Biochem, (Oct 20 1989), 185(1)79-84.
BERG88: Berg, JM, Proc Natl Acad Sci USA tl988), 85:99-102.
BETT88: Better, M, CP Chang, RR Robinson, and AH Horwitz, Science (1988), 240:1041-1043.
BOEK80: Boeke, JD, M Russel, and P Model, J Mol Biol (1980), 144:103-116.
CHAN79: Chang, CN, P Model, and G Blobel, Proc Natl Acad Sci USA (1979), 76:1251-1255.
CHOU74: Chou, PY, and GD Fasman, Biochemistry (1974),
13:(2)222-45.
C0~M87: Hemostasis and Thrombosis, Second Edition, Editors Colman, Hirsh, Marder, and Salzman, Published by Pippincott, Philadelphia, PA, 1987, ISBN 0-397-50679-1.
CREI84: Creighton, TE, Proteins: Structures and Molecular Princi~les, W H Freeman & Co, New York, 1984. -CWIR90: Cwirla, SE, EA Peters, RW Barrett, and WJ Dower, Proc Natl Acad Sci USA, ~August 1990), 87:6378-6382.
DELA88: de la Cruz, VF, AA ~al and TF McCutchan, J ~iol Chem, (1988), 263(9)4318-22.
DEV~90: Devlin, JJ, ~C Panganiban, and PE Devlin, Science, (27 July 1990), 249:404-406.
DICK83: Dickerson, RE, and I Geis, Hemoglobin: Structure Function. EvQlution. and Pathologv, The ~ejamin/Cummings Publishing Co, Menlo Park, CA, 1983.
DU~86: Dulbecco, R, US Patent 4,593,002, June 3, 1986.

.' ` '' ~``' '` ' `' ::

~ ~ 152 GAUS87: Gauss, P, KB Krassa, DS McPheeters, MA Nel~on, and L
Gold, Proc Natl Acad Sci USA (1987), 84:8515-19.
GIBS88: Gibson, TJ, JPM Postma, RS Brown, and P Argos, Protein Engineering (1988), 2(3)209-218.
GUAN91: Guan, KL and Dixon JE, Anal. Biochem, (1991), 192:
262-67.
HARD90: Hard, T, E Kellenbach, R Boelens, BA Maler, K
Dahlman, LP Freedman, J Carlstedt-Duke, XR Yamamoto, J-A Gustafsson, and R Kaptein, Science (13 ~uly 1990), 249:157-60.
HORV89: Horvat, S, B Grgas, N Raos, and VI Simeon, Int J
Peptide Protein Res (1989), 34:346-51.
INOU82: Inouye, H, W Barnes, and J Beckwith, J Bacteriol (1982), 149(2)434-439.
ITOK79: Ito, K, G Mandel, and W Wickner, Proc Natl Acad Sci USA (1979), 76:1199-1203.
JANA89: Janatova, J, KBM Reid, and AC Willis, Biochem (1989), 28:4754-61.
JANI85: Janin, J, and C Chothia, Methods in Enzymology (1985), 115~28)420-430.
KAPL78: Kaplan, DA, L Greenfield, and G Wilcox, in The Sin~le-Stranded DNA Pha~es, Denhardt, DT, D
Dressler, and DS Ray editors, Cold Spring Harbor Laboratory, 1978., p461-467.
KATZ90: Katz, B, and A~ Kossiakoff, Proteins, Struct, Funct, and Genet (1990), 7:343-57.
KIMH89: Kim, et al., Protein Engineering (1989), 2(1): 379-86.
KISH85: Kishore, R, and P Balaram, Biopolymers (1985), 24:2041-43.

. .

WO92/1;6~9 ~ 9, PCT/US92/01~39 .
KUHN85a: Kuhn, A, and W Wickner, J Biol Chem (1985), 260:15914-15918.
KUHN85b: Kuhn, A, and W Wickner, J Biol Chem (1985), 260:15907-15913.
KUHN87: Kuhn, A, Science (1987~, 238:1413-1415.
LISS85: Liss, LR, BL Johnson, and DB Oliver, J Bacteriol (1985), 164(2)925-8.
~OPE85a: Lopez, J, and RE Webster, J Bacteriol (1985), 163(3)1270-4.
LUIT85: Luiten, RGM, DG Putterman, JGG Schoenmakers, RNH
Konings, and LA Day, J Virology, (1985), 56(1)268-276.
LUIT87: Luiten, RGM, RIL Eggen, JGG Schoenmakers, and ~NH
Konings, DNA (1987), 6(2)129-37.
MARO80: Makowski, L, DLD Caspar, and DA Marvin, J Mol Biol (1980), 140:149-181. --MANI82: Maniatis, T, EF Frit~ch, and J Sambrook, Molecular Cloaina, Cold Spring Harbor ~aboratory, 1982.

MARK86: Marks, C~, M Vasser, P Ng, W Henzel, and S Anderson, J Biol Chem (1986), 261:7115-7118.
MARV78: Marvin, DA, in The Sinqle-Stranded DNA Phages, Denhardt, DT, D Dressler, and DS Ray editors, Cold Spring Harbor Laboratory, 1978., p583-603.
MATS89: Matsumura, M, WJ Becktel, M Levitt, and BW Matthews, Proc Natl Acad Sci USA (1989), 86:6562-6.
MCCA90: McCafferty, J, AD Griffiths, G Winter, and DJ
Chiswell, Nature, (6 Dec 1990), ~:552-4.
MESS77: Me~sing, J, B Gronenborn, B Muller-Hill, and P~ Hofschneider, Proc Natl Acad Sci USA (1977), 74:3642-6.
MæSS78: Mes~ing, J, and B Gronenborn, in The Single-Stranded ' .
.
.

WO92/1~679 PCT/US9'/~1~39 ~`3~ 154 DNA Phaqes, De~hardt, DT, D Dre881er, and DS Ray editors, Cold Spring Harbor Laboratory, 1978.,p449-453.
NICH88: Nicholson, H, WJ Becktel, and BW MAtthews, Nature (1988), 336:651-56.
NISH82: Nishiuchi, Y, and S Sakakibara, FE3S Lett (1982), ~:260-2.
NISH86: Nishiuchi, Y, ~ Kumagaye, Y Noda, TX Watanabe, and S
Sakakibara, Biopolymers, (1986), 25:S61-8.
OHKA81: Ohkawa, I, and RE Webster, J Biol Chem (19B1), 256:9951-9958.
OLIV9Oa: Olivera, BM, J Rivier, C Clark, CA Ramilo, GP
Corpuz, FC Abogadie, EE Mena, SR Woodward, DR
Hillyard, LJ Cruz, Science, (20 July 1990), 249:257-263.
PA~079: Pabo, CO, RT Sauer, JM Sturtevant, and M Ptashne, Proc Natl Acad Sci USA (1979), 76:1608-1612.
PARM88: Parmley, SF, and GP Smith, Gene (198B), 73:305-318.
PERR84: Perry, LJ, and R Wetzel, Science tl984), 226:555-7.
PERR86: Perry, W, and R Wetzel, Biochem (1986), 25:733-39.
POTE83: Poteete, AR, J Mol Biol (1983), 171:401-418.
RASC86: Rasched, I, and E Oberer, Microbiol Rev (1986) 50:401-427.
RASH84: Rashin, A, Biochemistry (1984), 23:5S18.
ROSE85: Rose, GD, Methods in Enzymololgy (1985), 115(29)430-440.
RUSS81: Russel, M, and P Model, Proc Natl Acad Sci USA
(1981), 78:1717-1721.
RUSS82: Russel and Model, Cell (1982), 28(1): 177-84.
SALI88: Sali, D, M Bycroft, and AR Fersht, Nature (1988), 335:74~-3.
SAM389: Sambrook, J, EF Fritsch, and T Maniatis, Molecular Cloninq A Laboratory Manual, Second Edition, Cold Spring Harbor ~aboratory, 1989.

W092/1~679 2 1 ~ ~ 3 ~ r ~ PCT/~S92tOtS39 SAUE86: Sauer, RT, K Hehir, RS Stearman, M~ Weiss, A Jeitler-Nilsson, EG Suchanek, and CO Pabo, Biochem (1986), 25:5992-98.
SCHA78: Schaller, X, E Beck, and M Takanami, in The Sinqle-Stranded DNA Phaqes, Denhardt, D.T., D. Dressler, and D.S. Ray editor~, Cold Spring ~arbor Laboratory, 1978., pl39-163.
SCHN86: Schnabel, E, W Schroeder, and G Reinhardt, Biol Chem Hoppe-Seyler (1986), 367:1167-76.
SCOT87a: Scott, MJ, CS Huckaby, I Kato, WJ Kohr, M Laskow3ki Jr., M-J Tsai and BW O~Malley, J Biol Chem (1987), ~(12)5899-5907.
SCOT90: Scott, JK, and GP Smith, Science, (27 July 1990), 249:386-390.
SMIT85: Smith GP, Science (1985), 228:1315-1317.
SUMM91: Summers, J. Cell Biochem. (1991), 45(1): 41-8.
VITA84: Vita, C, D Daizoppo, and A Fontana, Biochemistry (1984), 23:5512-5519.
wEBs7a: Web~ter, RE, and JS Cashman, in ~he Sinqle-Stranded DNA Pha~es, Denhardt, DT, D Dressler, and DS Ray editors, Cold Spring Harbor Laboratory, 1978., p557-569.
WELL86: Wells, JA, and DB Powers, J Biol Chem (1986), 261:6564-70.
YANI85: Yanisch-Perron, C, J Vieira, and J Messing, Gene, (1985), 33:103-119.

ZAFA88: Zafaralla, GC, C Ramilo, WR Gray, R Karlstrom, BM
Olivera, and LJ Cruz, Biochemictry, (1988), 27(18)7102-5.
ZIMM82: Zimmermann, R, C Watts, and W Wickner, J 3iol Chem (1982), 257:6529-6536.

~ . .. .

:

.

Claims (29)

156
1. A process for developing novel epitopes or binding proteins with a desired binding activity against a particular target material which comprises providing a library of phage which each displays on its surface, as a result of expression of a first phage gene, one or more copies of a particular chimeric coat protein, each chimeric coat protein comprising (a) a potential epitope, or a potential binding domain which is a mutant of a known protein domain foreign to said phage, as well as (b) at least a functional portion of a coat protein native to said phage, said library collectively displaying a plurality of potential epitopes or binding domains, contacting said library of phage with the target material, and separating the phage on the basis of their affinity for the target material, characterized in that said chimeric coat protein further comprises a linker peptide which is specifically cleavable by a site-specific protease, said linker peptide being positioned in between (a) said epitope or potential binding domain, and (b) said native coat protein sequence, whereby (a) may be freed from (b).
2. The method of claim 1 wherein the site, specific protease is Factor Xa, Factor XIa, kallikrein, thrombin, Factor XIIa, collagenase or enterokinase.
3. The method of claim 1 wherein, after said library of phage is contacted with said target material, (1) low affinity phage are removed, (2) high affinity phage still bound to said target material are released by cleavage of said chimeric coat protein at said linker by means of a site-specific protease, and the released high affinity phage are recovered.
4. A process for developing novel binding peptides or proteins with a desired binding activity against a particular target material which comprises providing a library of phage which each displays on its surface, as a result of expression of a first phage gene, one or more copies of a particular chimeric coat protein, each chimeric coat protein comprising a mutant of a known protein domain foreign to said phage, said library collectively displaying a plurality of potential binding domains, contacting said library of phage with the target material, and separating the phage on the basis of their affinity for the target material, characterized in that said potential binding domain has at least one intrachain covalent crosslink between a first amino acid position and a second amino acid position thereof, the amino acids at said first and second positions being invariant in all of the chimeric proteins displayed by said library, and where low affinity phage are removed from said target material first, and then high affinity phage are released or rendered more readily eluted from the target material by treating the phage with a reagent which cleaves the crosslink, preferably a reagent which does not kill the phage.
5. The method of any of claims 1.4 wherein the domain is a mini-protein of less than sixty amino acids, more preferably a micro-protein of less than forty amino acids.
6. The method of claim 4 wherein the crosslink is a disulfide bond and the amino acids at said first and second positions are cysteines.
7. The method of claim 6 wherein the reagent is dithiothreitol.
8. The method of any of claims 1.7 wherein the phage further comprises a second phage gene encoding the cognate wild-type coat protein of the phage.
9. A process for developing novel epitopes with a desired binding activity against a particular target material which comprises providing a library of phage which each displays on its surface, as a result of expression of a first phage gene, one or more copies of a particular chimeric coat protein, each chimeric coat protein comprising a potential epitope, said library collectively displaying a plurality of potential epitopes, contacting said library of phage with the target material, and separating the phage on the basis of their affinity for the target material, characterized in that the phage further comprises a second phage gene encoding the cognate wild-type coat protein of the phage.
10. A process for developing novel epitopes or binding proteins with a desired binding activity against a particular target material which comprises providing a library of phage which each displays on its surface, as a result of expression of a first phage gene, one or more copies of a particular chimeric coat protein, each chimeric coat protein comprising a potential epitope, or a potential binding domain which is a mutant of a known protein domain foreign to said phage, said library collectively displaying a plurality of potential epitopes or binding domains, contacting said library of phage with the target material, and separating the phage on the basis of their affinity for the target material, characterized in that the chimeric coat protein includes only an assemblable fragment of a coat protein of said phage, and not that portion of the coat protein which is responsible for pilus binding, and the phage also comprises a second phage gene encoding the cognate native coat protein of the phage.
11. A process for developing novel epitopes with a desired binding activity against a particular target material which comprises providing a library of phage which each displays on its surface, as a result of expression of a first phage gene, one or more copies of a particular chimeric coat protein, each chimeric coat protein comprising a potential epitope, said library collectively displaying a plurality of potential epitopes, contacting said library of phage with the target material, and separating the phage on the basis of their affinity for the target material, characterized in that the cognate wild-type coat protein of the phage is the major coat protein of the phage.
12. The method of claims 8-11 wherein the initiation condon of the second phage gene is a leucine.
13. The method of any of claims 1,12 wherein the first phage gene further comprises a cytoplasmic secretion signal sequence which codes for a signal peptide which directs the immediate expression product to the inner membrane of the bacterial host cell infected by said phage, where it is processed to remove said signal peptide, yielding a mature chimeric coat protein comprising the potential binding domain and at least a portion of a geneVIII.like protein of the phage, said chimeric protein being assembled with wild-type coat protein into the phage coat, wherein the secretion signal is encoded by a signal sequence selected from the group consisting of the signal sequences of the phoA, bla and geneIII genes.
14. A process for developing novel epitopes or binding proteins with a desired binding activity against a particular target material which comprises providing a library of phage which each displays on its surface, as a result of expression of a first phage gene, one or more copies of a particular chimeric coat protein, each chimeric coat protein comprising a potential epitope, or a potential binding domain which is a mutant of a known protein domain foreign to said phage, said library collectively displaying a plurality of potential epitopes or binding domains, contacting said library of phage with the target material, and separating the phage on the basis of their affinity for the target material, characterized in that the phage also comprises a second phage gene encoding the cognate native coat protein of the phage, and the initiation condon of the second phage gene is a Leucine condon.
15. The method of any of claims 1-14 wherein the differentiation among said plurality of different potential binding domains occurs through the controlled random variation of one or more predetermined amino acid positions of said known domain to randomly obtain at each said position an amino acid belonging to a predetermined set of two or more amino acids, the amino acids of said set occurring at said position in predetermined expected proportions.
16. The method of claim 15 wherein the differentiation among said potential binding domains of said library is limited to no more than about 20 predetermined amino acid residues of said sequence.
17. The method of claim 15 wherein, for each set, the ratio of the probability of occurrence of the most favored amino acid to that for the least favored amino acid is less than about 2.6.
18. The method of any of claims 1.17 wherein, for any potentially encoded potential binding domain, the probability that it will be displayed by at least one package in said population is at least 50%, more preferably at least 90%.
19. The method of any of claims 1.18 wherein said population is characterized by the display of at least 105 different potential binding domains.
20. The method of any of claims 1-19 wherein the initially chosen parental potential binding domain is selected from the group consisting of (a) binding domains of bovine pancreatic trypsin inhibitor, crambin, Cucurbita maxima trypsin inhibitor III, a heat.stable enterotoxin of Escherichia coli.
an alpha-, mu- or omega-conotoxin, apamin, charybdotoxin, secretory leukocyte protease inhibitor, cystatin, eglin, barley protease inhibitor, ovomucoid, T4 lysozyme, hen ege white lysozyme, ribonuclease, azurin, tumor necrosis factor, and CD4, and (b) domains at least substantially homologous with any of the foregoing domains which have a melting point of at least 50°C.
21. A process for developing novel epitopes with a desired affinity for a particular binding protein target material which comprises providing a library of phage which each displays on its surface, as a result of expression of a first phage gene, one or more copies of a particular chimeric coat protein, each chimeric coat protein comprising a potential epitope, or a potential binding domain which is a mutant of a known protein domain foreign to said phage, said library collectively displaying a plurality of potential epitopes or binding domains, contacting said library of phage with the target material, and separating the phage on the basis of their affinity for the binding protein target material, the differentiation among said plurality of different potential binding domains occurring through the controlled random variation, of one or more predetermined amino acid positions of said known domain to randomly obtain at each said position an amino acid belonging to a predetermined set of two or more amino acids; the amino acids of said set occurring at said position in predetermined expected proportions, and in substantially all sets the ratio of the frequency of occurrence of the most favored amino acid to that for the least favored amino acid is less than 2.6, characterized in that, at at least one such position, the predetermined set consists of less than all twenty different genetically encodable amino acids, but includes three or more of the classes of amino acids, said classes being acidic (Asp, Glu), basic (Arg, Lys), aliphatic hydrophobic (Leu, Ile, Ala, Cys, Met, Val, Thr), aromatic hydrophobic (His, Tyr, Phe, Trp), neutral hydrophilic (ser, Asn, Gln), neutral flexible (Gly) and neutral rigid (Pro).
22. The method of claim 21 in which at least one variable amino acid position is encoded by a simply variegated condon selected from the group consisting of NNT, NNG, RNG, RMG, VNT, RRS, SNT, and VHG.
23. The method of claim 21 wherein none of the variable amino acid positions is encoded by a simply variegated condon selected from the group consisting of NNN, NNK and NNS.
24. The method of claim 21 in which at least one variable amino acid position is encoded by a complexly variegated condon.
25. A library of display phage which each displays on its surface, as a result of expression of a first phage gene, one or more copies of a particular chimeric coat protein, each chimeric coat protein comprising a potential epitope, or a potential binding domain which is a mutant of a known protein domain foreign to said phage, said library collectively displaying a plurality of potential epitopes or binding domains, wherein the differentiation among said plurality of different potential binding domains occurs through the at least partially random variation of one or more predetermined amino acid positions of said known domain to randomly obtain at each said position an amino acid belonging to a predetermined set of two or more amino acids, the amino acids of said set occurring at said position in predetermined expected proportions, and in substantially all sets the ratio of the frequency of occurrence of the most favored amino acid to that for the least favored amino acid is less than 2.6, characterized in that, at at least one such position, the predetermined set consists of less than all twenty different genetically encodable amino acids, but includes two or more of the classes of genetically encodable amino acids.
26. a library of display phage which each displays on its surface, as a result of expression of a first phage gene, one or more copies of a particular chimeric coat protein, each chimeric coat protein comprising (a) a potential epitope, or a potential binding domain which is a mutant of a known protein domain foreign to said phage, as well as (b) at least a functional portion of a coat protein native to said phage, said library collectively displaying a plurality of potential epitopes or binding domains, wherein said chimeric coat protein further comprises a linker peptide which is specifically cleavable by said site-specific protease, said linker peptide being positioned inbetween (a) said epitope or potential binding domain, and (b) said native coat protein sequence, whereby (a) may be freed from (b).
27. The method of any of claims 21-24 in which at least one variable position the substitution set includes all classes of genetically encodable amino acid.
28. The method of any of claims 21-24 in which the known protein domain does not bind or only weakly binds the target, as indicated by a dissociation constant of greater then 106 motes/liter for the complex of the domain with the target.
29. The method of claim 21 wherein for at least one variable condon, the substitution set excludes cysteine.
CA002105303A 1991-03-01 1992-02-28 Display phage Abandoned CA2105303A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US07/664,989 US5223409A (en) 1988-09-02 1991-03-01 Directed evolution of novel binding proteins
US664,989 1991-03-01

Publications (1)

Publication Number Publication Date
CA2105303A1 true CA2105303A1 (en) 1992-09-02

Family

ID=24668274

Family Applications (2)

Application Number Title Priority Date Filing Date
CA002105300A Expired - Lifetime CA2105300C (en) 1991-03-01 1992-02-27 Process for the development of binding mini-proteins
CA002105303A Abandoned CA2105303A1 (en) 1991-03-01 1992-02-28 Display phage

Family Applications Before (1)

Application Number Title Priority Date Filing Date
CA002105300A Expired - Lifetime CA2105300C (en) 1991-03-01 1992-02-27 Process for the development of binding mini-proteins

Country Status (10)

Country Link
US (1) US7893007B2 (en)
EP (6) EP1279731B1 (en)
JP (6) JP4146512B2 (en)
AT (4) ATE439435T1 (en)
AU (2) AU1545692A (en)
CA (2) CA2105300C (en)
DE (4) DE69233769D1 (en)
DK (4) DK1279731T3 (en)
ES (4) ES2287206T3 (en)
WO (2) WO1992015677A1 (en)

Families Citing this family (641)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5223409A (en) 1988-09-02 1993-06-29 Protein Engineering Corp. Directed evolution of novel binding proteins
US7413537B2 (en) 1989-09-01 2008-08-19 Dyax Corp. Directed evolution of disulfide-bonded micro-proteins
US5747334A (en) * 1990-02-15 1998-05-05 The University Of North Carolina At Chapel Hill Random peptide library
US5498538A (en) * 1990-02-15 1996-03-12 The University Of North Carolina At Chapel Hill Totally synthetic affinity reagents
US5723286A (en) * 1990-06-20 1998-03-03 Affymax Technologies N.V. Peptide library and screening systems
DK1279731T3 (en) 1991-03-01 2007-09-24 Dyax Corp Process for the development of binding mini-proteins
US5733731A (en) * 1991-10-16 1998-03-31 Affymax Technologies N.V. Peptide library and screening method
US5270170A (en) * 1991-10-16 1993-12-14 Affymax Technologies N.V. Peptide library and screening method
US6610656B1 (en) 1993-12-30 2003-08-26 President And Fellows Of Harvard College Method of promoting chondrocyte differentiation with hedgehog related polypeptides
US6165747A (en) 1993-12-30 2000-12-26 President & Fellows Of Harvard College Nucleic acids encoding hedgehog proteins
US6884775B1 (en) 1993-12-30 2005-04-26 President And Fellows Of Harvard College Methods and compositions for regulating skeletogenic formation
US6271363B1 (en) 1993-12-30 2001-08-07 President & Fellows Of Harvard College Nucleic acids encoding hedgehog proteins
US6261786B1 (en) 1993-12-30 2001-07-17 Imperial Cancer Res. Technology Screening assays for hedgehog agonists and antagonists
US20030186357A1 (en) 1993-12-30 2003-10-02 Philip W. Ingham Vertebrate embryonic pattern-inducing proteins, and uses related thereto
US7060450B1 (en) 1993-12-30 2006-06-13 President And Fellows Of Harvard College Screening assays for agonists and antagonists of the hedgehog signaling pathway
PT1231268E (en) 1994-01-31 2005-11-30 Univ Boston BANKS OF POLYCLONE ANTIBODIES
US5565327A (en) * 1994-03-25 1996-10-15 Duke University Methods of diagnosing parasitic infections and of testing drug susceptibility of parasites
EP0699750A1 (en) * 1994-06-07 1996-03-06 Gesellschaft für biotechnologische Forschung mbH (GBF) A collection of phagemids, a collection of Escherichia coli cells carrying the phagemids, a collection of phagemid particles produced from said collection and phagemid particles obtained according to the process
US6576236B1 (en) 1994-07-01 2003-06-10 Dana Farber Cancer Institute Methods for stimulating T cell responses by manipulating a common cytokine receptor γ chain
DE69526665T2 (en) * 1994-09-21 2002-11-21 Cytogen Corp ANTI-BINDING PEPTIDES (ABTIDES) FROM PEPTIDE LIBRARIES
US5885577A (en) * 1994-09-21 1999-03-23 Cytogen Corporation Antigen binding peptides (abtides) from peptide libraries
US6475806B1 (en) 1995-06-07 2002-11-05 Praecis Pharmaceuticals, Inc. Anchor libraries and identification of peptide binding sequences
US6090382A (en) 1996-02-09 2000-07-18 Basf Aktiengesellschaft Human antibodies that bind human TNFα
AU773394B2 (en) * 1996-10-08 2004-05-27 U-Bisys B.V. Methods and means for selecting peptides and proteins having specific affinity for a target
ATE230850T1 (en) * 1996-10-08 2003-01-15 Bisys B V U METHOD AND MEANS FOR SELECTING PEPTIDES AND PROTEINS WITH SPECIFIC AFFINITY FOR A TARGET MOLECULE
SE9700291D0 (en) * 1997-01-31 1997-01-31 Pharmacia & Upjohn Ab Selection method and prodcts resulting therefrom
US6121416A (en) 1997-04-04 2000-09-19 Genentech, Inc. Insulin-like growth factor agonist molecules
US6420518B1 (en) 1997-04-04 2002-07-16 Genetech, Inc. Insulin-like growth factor agonist molecules
EP0975755B2 (en) 1997-04-16 2011-08-03 Millennium Pharmaceuticals, Inc. Crsp protein (cysteine-rich secreted proteins), nucleic acid molecules encoding them and uses therefor
AU8173098A (en) 1997-06-27 1999-01-19 Ontogeny, Inc. Neuroprotective methods and reagents
US7144997B2 (en) 1997-07-24 2006-12-05 Curis, Inc. Vertebrate embryonic patterning-inducing proteins, compositions and uses related therto
US6639051B2 (en) 1997-10-20 2003-10-28 Curis, Inc. Regulation of epithelial tissue by hedgehog-like polypeptides, and formulations and uses related thereto
US7163682B2 (en) 1998-04-13 2007-01-16 The Forsyth Institute Glucan binding protein and glucosyltransferase immunogens
US7056517B2 (en) 1998-04-13 2006-06-06 The Forsyth Institute Glucosyltransferase immunogens
WO1999058655A2 (en) * 1998-05-13 1999-11-18 Diversys Limited Phage display selection system for folded proteins
WO2000017336A1 (en) * 1998-09-19 2000-03-30 Sang Jun Lee Dna cassette encoding a multimer of a biologically active peptide and a cleavable linker attached thereto and process for preparing the biologically active peptide
US6420110B1 (en) 1998-10-19 2002-07-16 Gpc Biotech, Inc. Methods and reagents for isolating biologically active peptides
US6884770B1 (en) 1998-11-06 2005-04-26 Curis, Inc. Methods and compositions for treating or preventing peripheral neuropathies
US20040009535A1 (en) 1998-11-27 2004-01-15 Celltech R&D, Inc. Compositions and methods for increasing bone mineralization
DE69942782D1 (en) 1998-11-27 2010-10-28 Ucb Sa Compositions and methods for increasing bone mineralization
IL143866A0 (en) 1999-01-06 2002-04-21 Genentech Inc Insulin-like growth factor (igf) i mutant variants
AU3382800A (en) 1999-02-26 2000-09-14 Millennium Pharmaceuticals, Inc. Secreted proteins and uses thereof
WO2000057183A1 (en) * 1999-03-23 2000-09-28 Biovation Limited Protein isolation and analysis
US6864235B1 (en) 1999-04-01 2005-03-08 Eva A. Turley Compositions and methods for treating cellular response to injury and other proliferating cell disorders regulated by hyaladherin and hyaluronans
DE19915057A1 (en) 1999-04-01 2000-10-19 Forschungszentrum Borstel Monoclonal antibodies to the human Mcm3 protein, process for their preparation and their use
US6911429B2 (en) 1999-04-01 2005-06-28 Transition Therapeutics Inc. Compositions and methods for treating cellular response to injury and other proliferating cell disorders regulated by hyaladherin and hyaluronans
GB9908195D0 (en) 1999-04-09 1999-06-02 Microbiological Res Authority Treatment of intracellular infection
US20040001826A1 (en) 1999-06-30 2004-01-01 Millennium Pharmaceuticals, Inc. Glycoprotein VI and uses thereof
US7291714B1 (en) 1999-06-30 2007-11-06 Millennium Pharmaceuticals, Inc. Glycoprotein VI and uses thereof
PT1210428E (en) 1999-08-23 2015-07-21 Genetics Inst Llc Pd-1, a receptor for b7-4, and uses therefor
US8168178B2 (en) 1999-11-30 2012-05-01 Curis, Inc. Methods and compositions for regulating lymphocyte activity
US6951839B1 (en) 1999-11-30 2005-10-04 Curis, Inc. Methods and compositions for regulating lymphocyte activity
JP2003518942A (en) 1999-12-30 2003-06-17 プレジデント・アンド・フェローズ・オブ・ハーバード・カレッジ Methods and compositions relating to modulating hepatocyte growth, plasma cell differentiation, or T cell subset activity by modulation of XBP-1 activity
KR20110032012A (en) 2000-02-10 2011-03-29 아보트 러보러터리즈 Antibodies that bind human interleukin-18 and methods of making and using
US7351540B1 (en) 2000-03-17 2008-04-01 Merck Patent Gmbh Protein isolation and analysis
DK1282437T3 (en) 2000-05-16 2008-06-30 Genentech Inc Treatment of cartilage disorders
EP1714661A3 (en) 2000-05-19 2012-03-14 The Center for Blood Research, INC. Methods for diagnosing and treating hemostatic disorders by modulating p-selectin activity
EP1286682B1 (en) 2000-06-08 2010-03-03 Immune Disease Institute, Inc. Methods and compositions for inhibiting immunoglobulin-mediated-reperfusion injury
CA2408630A1 (en) 2000-06-19 2001-12-27 Dyax Corp. Novel enterokinase cleavage sequences
AU7309601A (en) 2000-06-28 2002-01-08 Genetics Inst Pd-l2 molecules: novel pd-1 ligands and uses therefor
AU2001277648A1 (en) 2000-07-14 2002-04-15 Cropdesign N.V. Plant cyclin-dependent kinase inhibitors
JP5113314B2 (en) 2000-09-01 2013-01-09 ザ センター フォー ブラッド リサーチ インク Modified polypeptide stabilized in desired conformation and method for producing the polypeptide
EP1339427A4 (en) 2000-11-01 2004-09-15 Elusys Therapeutics Inc Method of producing biospecific molecules by protein trans-splicing
AU2888602A (en) 2000-11-03 2002-05-15 Pbl Biomedical Lab Interferons, uses and compositions related thereto
EP1356108A2 (en) 2000-11-28 2003-10-29 Wyeth Expression analysis of fkbp nucleic acids and polypeptides useful in the diagnosis and treatment of prostate cancer
HUP0303905A3 (en) 2000-11-28 2005-11-28 Wyeth Corp Expression analysis of kiaa nucleic acids and polypeptides useful in the diagnosis and treatment of prostate cancer
US9249229B2 (en) 2000-12-08 2016-02-02 Alexion Pharmaceuticals, Inc. Polypeptides and antibodies derived from chronic lymphocytic leukemia cells and uses thereof
AU2002246632B2 (en) 2000-12-08 2007-04-05 Alexion Pharmaceuticals, Inc. Chronic lymphocytic leukemia cell line and its use for producing an antibody
US7408041B2 (en) 2000-12-08 2008-08-05 Alexion Pharmaceuticals, Inc. Polypeptides and antibodies derived from chronic lymphocytic leukemia cells and uses thereof
US20060057651A1 (en) 2000-12-08 2006-03-16 Bowdish Katherine S Polypeptides and antibodies derived from chronic lymphocytic leukemia cells and uses thereof
AR032028A1 (en) 2001-01-05 2003-10-22 Pfizer ANTIBODIES AGAINST THE RECEIVER OF THE SIMILAR TO INSULIN GROWTH FACTOR
EP1377664A2 (en) 2001-02-23 2004-01-07 DSM IP Assets B.V. Genes encoding proteolytic enzymes from aspargilli
WO2002070662A2 (en) 2001-03-02 2002-09-12 Gpc Biotech Ag Three hybrid assay system
JP2005508608A (en) 2001-03-22 2005-04-07 アボット ゲーエムベーハー ウント カンパニー カーゲー Transgenic animals expressing antibodies specific to the gene of interest and uses thereof
CA2442066C (en) 2001-04-02 2005-11-01 Wyeth Pd-1, a receptor for b7-4, and uses therefor
KR100886095B1 (en) 2001-04-16 2009-02-27 와이어쓰 홀딩스 코포레이션 Novel Streptococcus pneumoniae open reading frames encoding polypeptide antigens and a composition comprising the same
DE60228477D1 (en) 2001-05-08 2008-10-02 Darwin Molecular Corp PROCESS FOR REGULATING THE IMMUNE FUNCTION IN PRIMATES USING THE FOXP3 PROTEIN
CA2817619A1 (en) 2001-06-08 2002-12-08 Abbott Laboratories (Bermuda) Ltd. Methods of administering anti-tnf.alpha. antibodies
MXPA03011890A (en) 2001-06-22 2004-06-03 Du Pont Defensin polynucleotides and methods of use.
US6833441B2 (en) 2001-08-01 2004-12-21 Abmaxis, Inc. Compositions and methods for generating chimeric heteromultimers
US20040142325A1 (en) 2001-09-14 2004-07-22 Liat Mintz Methods and systems for annotating biomolecular sequences
US7175983B2 (en) 2001-11-02 2007-02-13 Abmaxis, Inc. Adapter-directed display systems
AR039067A1 (en) 2001-11-09 2005-02-09 Pfizer Prod Inc ANTIBODIES FOR CD40
ES2337989T3 (en) 2001-12-18 2010-05-03 Endocube Sas NEW PROTEINS ASSOCIATED WITH THE DEATH OF THE THAP FAMILY AND RELATED PAR4 ROUTES INVOLVED IN THE CONTROL OF APOPTOSIS.
US7858297B2 (en) 2001-12-18 2010-12-28 Centre National De La Recherche Scientifique Cnrs Chemokine-binding protein and methods of use
EP1463807A4 (en) 2001-12-19 2006-04-12 Bristol Myers Squibb Co Pichia pastoris formate dehydrogenase and uses therefor
EP1572149A2 (en) 2002-03-07 2005-09-14 The Forsyth Institute Immunogenicity of glucan binding protein
US7745192B2 (en) 2002-04-03 2010-06-29 Venomics Pty Limited Prothrombin activating protein
US20030206898A1 (en) 2002-04-26 2003-11-06 Steven Fischkoff Use of anti-TNFalpha antibodies and another drug
PL373887A1 (en) 2002-05-21 2005-09-19 Dsm Ip Assets B.V. Novel phospholipases and uses thereof
USRE47770E1 (en) 2002-07-18 2019-12-17 Merus N.V. Recombinant production of mixtures of antibodies
EP1523496B1 (en) 2002-07-18 2011-06-29 Merus B.V. Recombinant production of mixtures of antibodies
DK1944322T3 (en) 2002-07-19 2015-06-22 Abbvie Biotechnology Ltd Treatment of TNFalfa-related diseases
US7250551B2 (en) 2002-07-24 2007-07-31 President And Fellows Of Harvard College Transgenic mice expressing inducible human p25
EA200500330A1 (en) 2002-08-10 2006-06-30 Йейл Юниверсити ANTAGONISTS NOGO RECEPTORS
ES2527924T3 (en) 2002-08-19 2015-02-02 Dsm Ip Assets B.V. New lipases and their uses
EP2311980A1 (en) 2002-08-20 2011-04-20 Millennium Pharmaceuticals, Inc. Compositions, kits, and methods for identification, assessment, prevention, and therapy of cervical cancer
AU2003250518A1 (en) 2002-08-20 2004-03-11 Yeda Research And Development Co. Ltd. Akap84 and its use for visualization of biological structures
WO2004021994A2 (en) 2002-09-04 2004-03-18 Biopolymer Engineering, Inc. Cancer therapy using whole glucan particles and antibodies
EP2258724A1 (en) 2002-11-21 2010-12-08 Celltech R & D, Inc. Modulating immune responses using multimerized anti-CD83 antibodies
EP2290107A1 (en) 2003-01-07 2011-03-02 Dyax Corporation Kunitz domain library
DE10303974A1 (en) 2003-01-31 2004-08-05 Abbott Gmbh & Co. Kg Amyloid β (1-42) oligomers, process for their preparation and their use
US20040180387A1 (en) 2003-03-13 2004-09-16 Fujirebio Diagnostics, Inc. Detection of urinary mesothelin-/megakaryocyte potentiating factor-related peptides for assessment of ovarian cancer
US20050014932A1 (en) 2003-05-15 2005-01-20 Iogenetics, Llc Targeted biocides
WO2004106375A1 (en) 2003-05-30 2004-12-09 Merus Biopharmaceuticals B.V. I.O. Fab library for the preparation of anti vegf and anti rabies virus fabs
US20100069614A1 (en) 2008-06-27 2010-03-18 Merus B.V. Antibody producing non-human mammals
CA2542232A1 (en) 2003-06-09 2005-01-20 Alnylam Pharmaceuticals, Inc. Method for treating neurodegenerative disease by inhibiting alpha-synuclein
HN2004000285A (en) 2003-08-04 2006-04-27 Pfizer Prod Inc ANTIBODIES DIRECTED TO c-MET
AR045563A1 (en) 2003-09-10 2005-11-02 Warner Lambert Co ANTIBODIES DIRECTED TO M-CSF
MXPA06003402A (en) 2003-10-07 2006-06-27 Millennium Pharm Inc Nucleic acid molecules and proteins for the identification, assessment, prevention, and therapy of ovarian cancer.
US20050100965A1 (en) 2003-11-12 2005-05-12 Tariq Ghayur IL-18 binding proteins
MX350383B (en) 2004-01-09 2017-09-04 Pfizer ANTIBODIES TO MAdCAM.
JP5912211B2 (en) 2004-01-20 2016-04-27 メルス ビー.ヴィー. Mixture of binding proteins
EP1714151A4 (en) 2004-01-21 2009-06-10 Fujirebio America Inc Detection of mesothelin-/megakaryocyte potentiating factor-related peptides in peritoneal fluid for assessment of the peritoneum and the peritoneal cavity
CA2560066A1 (en) 2004-03-01 2005-09-15 The Cbr Institute For Biomedical Research, Inc. Natural igm antibodies and inhibitors thereof
KR100882249B1 (en) 2004-03-24 2009-02-06 트리패스 이미징, 인코포레이티드 Methods and compositions for the detection of cervical disease
TW201705980A (en) 2004-04-09 2017-02-16 艾伯維生物技術有限責任公司 Multiple-variable dose regimen for treating TNF[alpha]-related disorders
CN101133157A (en) 2004-06-03 2008-02-27 阿什洛米克斯控股有限公司 Agents and methods for diagnosing stress
GB0414054D0 (en) 2004-06-23 2004-07-28 Owen Mumford Ltd Improvements relating to automatic injection devices
WO2006008639A1 (en) 2004-07-16 2006-01-26 Pfizer Products Inc. Combination treatment for non-hematologic malignancies using an anti-igf-1r antibody
US7342093B2 (en) 2004-07-23 2008-03-11 University Of Massachusetts Compounds that inhibit Hsp90 protein-protein interactions with IAP proteins
GT200600031A (en) 2005-01-28 2006-08-29 ANTI-BETA ANTIBODY FORMULATION
EA015584B1 (en) 2005-03-23 2011-10-31 Генмаб А/С Antibody to human cd38 and use thereof
AR053067A1 (en) 2005-04-25 2007-04-18 Pfizer ANTIBODIES AGAINST MYOSTATIN
BRPI0608096A2 (en) 2005-04-26 2009-11-10 Pfizer p-cadherin antibodies
US7595380B2 (en) 2005-04-27 2009-09-29 Tripath Imaging, Inc. Monoclonal antibodies and methods for their use in the detection of cervical disease
JP5757495B2 (en) 2005-05-16 2015-07-29 アッヴィ バイオテクノロジー リミテッド Use of TNF inhibitors for the treatment of erosive polyarthritis
EP3805245A1 (en) 2005-05-17 2021-04-14 University of Connecticut Compositions and methods for immunomodulation in an organism
HUE030701T2 (en) 2005-05-27 2017-05-29 Biogen Ma Inc Tweak binding antibodies
EP2388274A1 (en) 2005-06-17 2011-11-23 Janssen Alzheimer Immunotherapy Methods of purifying anti A Beta antibodies
JP5142458B2 (en) 2005-06-30 2013-02-13 キヤノン株式会社 Target substance capture molecule, target substance capture element, target substance detection apparatus and kit using these, and target substance detection method
NZ596992A (en) 2005-06-30 2013-07-26 Abbott Lab Il-12/p40 binding proteins
WO2007006091A1 (en) 2005-07-07 2007-01-18 Athlomics Pty Ltd Polynucleotide marker genes and their expression, for diagnosis of endotoxemia
GB2428293A (en) * 2005-07-13 2007-01-24 Domantis Ltd Phage display libraries
WO2007024715A2 (en) 2005-08-19 2007-03-01 Abbott Laboratories Dual variable domain immunoglobin and uses thereof
US7612181B2 (en) 2005-08-19 2009-11-03 Abbott Laboratories Dual variable domain immunoglobulin and uses thereof
US20070041905A1 (en) 2005-08-19 2007-02-22 Hoffman Rebecca S Method of treating depression using a TNF-alpha antibody
EP2500358A3 (en) 2005-08-19 2012-10-17 Abbott Laboratories Dual variable domain immunoglobulin and uses thereof
PL2960253T3 (en) 2005-09-07 2018-11-30 Amgen Fremont Inc. Human monoclonal antibodies to activin receptor-like kinase-1
CN101277974A (en) 2005-09-30 2008-10-01 阿伯特有限及两合公司 Binding domains of proteins of the repulsive guidance molecule (RGM) protein family and functional fragments thereof, and their use
EP2862867A3 (en) 2005-10-25 2015-08-05 The Johns Hopkins University Methods and compositions for the treatment of Marfan syndrome and associated disorders
BRPI0618085A2 (en) 2005-11-01 2011-08-16 Abbott Biotech Ltd Processes and kits for diagnosis of ankylosing spondylitis using biomarkers
EP2365334A1 (en) 2005-11-14 2011-09-14 MetaMol Theranostics, LLC Peptide sequence that promotes tumor invasion
MX2008006945A (en) 2005-11-30 2008-10-02 Massachusetts Inst Technology Pathogen detection biosensor.
PT1976877E (en) 2005-11-30 2014-04-29 Abbvie Inc Monoclonal antibodies against amyloid beta protein and uses thereof
EP1954718B1 (en) 2005-11-30 2014-09-03 AbbVie Inc. Anti-a globulomer antibodies, antigen-binding moieties thereof, corresponding hybridomas, nucleic acids, vectors, host cells, methods of producing said antibodies, compositions comprising said antibodies, uses of said antibodies and methods of using said antibodies
US20070264687A1 (en) 2005-12-15 2007-11-15 Min-Yuan Chou Recombinant triplex scaffold-based polypeptides
US8014957B2 (en) 2005-12-15 2011-09-06 Fred Hutchinson Cancer Research Center Genes associated with progression and response in chronic myeloid leukemia and uses thereof
US10183986B2 (en) 2005-12-15 2019-01-22 Industrial Technology Research Institute Trimeric collagen scaffold antibodies
US7632498B2 (en) 2005-12-19 2009-12-15 Tripath Imaging, Inc. MCM6 and MCM7 monoclonal antibodies and methods for their use in the detection of cervical disease
EP1803814A1 (en) * 2005-12-27 2007-07-04 SIGMA-TAU Industrie Farmaceutiche Riunite S.p.A. Method of improving the antibody selection capacity in phage-display library
PL2463305T3 (en) 2006-01-12 2017-02-28 Alexion Pharmaceuticals, Inc. Antibodies to OX-2/CD200 and uses thereof
KR20080094803A (en) 2006-01-27 2008-10-24 트리패스 이미징, 인코포레이티드 Methods for identifying patients with an increased likelihood of having ovarian cancer and compositions therefor
PT1981902E (en) 2006-01-27 2015-11-02 Biogen Ma Inc Nogo receptor antagonists
TW200744634A (en) 2006-02-21 2007-12-16 Wyeth Corp Methods of using antibodies against human IL-22
TWI417301B (en) 2006-02-21 2013-12-01 Wyeth Corp Antibodies against human il-22 and uses therefor
JP2009529915A (en) 2006-03-20 2009-08-27 ゾーマ テクノロジー リミテッド Human antibodies and methods specific for gastrin substances
CA2647029A1 (en) 2006-04-05 2007-10-18 Abbott Biotechnology Ltd. Antibody purification
CA2564435A1 (en) 2006-04-10 2007-10-10 Abbott Biotechnology Ltd. Methods for monitoring and treating intestinal disorders
EP2708242A3 (en) 2006-04-10 2014-03-26 Abbott Biotechnology Ltd Uses and compositions for treatment of ankylosing spondylitis
EP2666472A3 (en) 2006-04-10 2014-04-02 Abbott Biotechnology Ltd Uses and compositions for treatment of psoriatic arthritis
US10522240B2 (en) 2006-05-03 2019-12-31 Population Bio, Inc. Evaluating genetic disorders
US7702468B2 (en) 2006-05-03 2010-04-20 Population Diagnostics, Inc. Evaluating genetic disorders
EP2043711A4 (en) 2006-06-30 2017-08-30 AbbVie Biotechnology Ltd Automatic injection device
US8211428B2 (en) 2006-07-05 2012-07-03 Torrey Pines Institute For Molecular Studies Protease screening methods and proteases identified thereby
WO2008005529A2 (en) * 2006-07-07 2008-01-10 The Trustees Columbia University In The City Of New York Cell-mediated directed evolution
DK2511301T3 (en) 2006-08-04 2018-03-12 Medimmune Ltd HUMAN ANTIBODIES AGAINST ERBB 2
CA2914170C (en) 2006-09-08 2018-10-30 Abbvie Bahamas Ltd. Interleukin-13 binding proteins
US20080124355A1 (en) 2006-09-22 2008-05-29 David Gordon Bermudes Live bacterial vaccines for viral infection prophylaxis or treatment
CA2664740C (en) 2006-09-26 2021-11-16 Genmab A/S Combination treatment of cd38-expressing tumors
US7744890B2 (en) 2006-10-12 2010-06-29 Wyeth Llc Methods and compositions with reduced opalescence
JP5401319B2 (en) 2006-11-03 2014-01-29 ワイス・エルエルシー Glycolytic inhibitors in cell culture
NZ576878A (en) 2006-11-15 2011-10-28 Functional Genetics Inc Anti-TSG101 antibody (ATCC deposit PTA-9611) and its use for treatment of viral infection
US7488807B2 (en) 2006-11-22 2009-02-10 3M Innovative Properties Company Antibody with protein A selectivity
US8455626B2 (en) 2006-11-30 2013-06-04 Abbott Laboratories Aβ conformer selective anti-aβ globulomer monoclonal antibodies
EA017529B1 (en) 2006-12-05 2013-01-30 Декоуд Дженетикс Ехф. Genetic markers for risk management of cardiac arrhythmia
CA2673592C (en) 2006-12-20 2014-03-25 Xoma Technology Ltd. Methods for the treatment of il-1.beta. related diseases
CA3045637A1 (en) 2006-12-27 2008-07-10 Emory University Compositions and methods for the treatment of infections and tumors
SG193026A1 (en) 2007-02-07 2013-09-30 Decode Genetics Ehf Genetic variants contributing to risk of prostate cancer
MX2009008878A (en) 2007-02-21 2009-08-28 Decode Genetics Ehf Genetic susceptibility variants associated with cardiovascular disease.
US20100311767A1 (en) 2007-02-27 2010-12-09 Abbott Gmbh & Co. Kg Method for the treatment of amyloidoses
ATE544854T1 (en) 2007-03-22 2012-02-15 Heptares Therapeutics Ltd MUTANT G-PROTEIN-COUPLED RECEPTORS AND SELECTION METHODS THEREOF
KR20100018499A (en) 2007-04-02 2010-02-17 암젠 프레몬트 인코포레이티드 Anti-ige antibodies
MX2009010997A (en) 2007-04-13 2009-11-02 Catalyst Biosciences Inc Modified factor vii polypetides and uses thereof.
WO2008146309A2 (en) 2007-05-25 2008-12-04 Decode Genetics Ehf. Genetic variants on chr 5pl2 and 10q26 as markers for use in breast cancer risk assessment, diagnosis, prognosis and treatment
EP2171451A4 (en) 2007-06-11 2011-12-07 Abbott Biotech Ltd Methods for treating juvenile idiopathic arthritis
ES2528167T3 (en) 2007-06-15 2015-02-04 Deutsches Krebsforschungszentrum Stiftung des öffentlichen Rechts Tumor treatment using specific anti-L1 antibody.
BRPI0814645A2 (en) 2007-07-25 2015-01-27 Alexion Pharma Inc METHODS AND COMPOSITIONS FOR TREATING AUTOIMMUNE DISEASE.
US8697360B2 (en) 2007-11-30 2014-04-15 Decode Genetics Ehf. Genetic variants on CHR 11Q and 6Q as markers for prostate and colorectal cancer predisposition
GB0724051D0 (en) 2007-12-08 2008-01-16 Medical Res Council Mutant proteins and methods for producing them
PT2391650E (en) 2007-12-20 2015-01-14 Xoma Us Llc Methods for the treatment of gout
GB0724860D0 (en) 2007-12-20 2008-01-30 Heptares Therapeutics Ltd Screening
PT2237803E (en) 2007-12-28 2015-10-16 Prothena Biosciences Ltd Treatment and prophylaxis of amyloidosis
GB0802474D0 (en) 2008-02-11 2008-03-19 Heptares Therapeutics Ltd Mutant proteins and methods for selecting them
NZ587903A (en) 2008-02-14 2013-05-31 Decode Genetics Ehf Susceptibility for lung cancer using the polymorphic marker rs1051730
EP2271366A4 (en) 2008-02-28 2012-06-20 3M Innovative Properties Co Antibodies to clostridium difficile spores and uses thereof
US8962803B2 (en) 2008-02-29 2015-02-24 AbbVie Deutschland GmbH & Co. KG Antibodies against the RGM A protein and uses thereof
WO2009109572A2 (en) * 2008-03-03 2009-09-11 Ablynx Nv Monovalent phage display of single variable domains
EP2274450A2 (en) 2008-04-01 2011-01-19 Decode Genetics EHF Susceptibility variants for peripheral arterial disease and abdominal aortic aneurysm
BR122021026834B1 (en) 2008-04-25 2022-11-08 Takeda Pharmaceutical Company Limited ISOLATED ANTIBODIES BINDING FCRN, PHARMACEUTICAL COMPOSITION COMPRISING THEMSELVES AND METHODS TO DETECT AND MODULATE FCRN
NZ588554A (en) 2008-04-29 2013-03-28 Abbott Lab Dual variable domain immunoglobulins and uses thereof
SG188142A1 (en) 2008-05-09 2013-03-28 Abbott Gmbh & Co Kg Antibodies to receptor of advanced glycation end products (rage) and uses thereof
CN102112494A (en) 2008-06-03 2011-06-29 雅培制药有限公司 Dual variable domain immunoglobulins and uses thereof
AR072001A1 (en) 2008-06-03 2010-07-28 Abbott Lab IMMUNOGLOBULIN WITH DUAL VARIABLE DOMAIN AND USES OF THE SAME
EP2313525A2 (en) 2008-07-07 2011-04-27 Decode Genetics EHF Genetic variants for breast cancer risk assessment
TW201014602A (en) 2008-07-08 2010-04-16 Abbott Lab Prostaglandin E2 binding proteins and uses thereof
WO2010006060A2 (en) 2008-07-08 2010-01-14 Abbott Laboratories Prostaglandin e2 dual variable domain immunoglobulins and uses thereof
CA2729961C (en) 2008-07-09 2018-05-01 Biogen Idec Ma Inc. Li113, li62 variant co2, anti-lingo antibodies
EP3199630B1 (en) 2008-09-05 2019-05-08 President and Fellows of Harvard College Continuous directed evolution of proteins and nucleic acids
EP2344180A2 (en) 2008-09-23 2011-07-20 Wyeth LLC Methods for predicting production of activating signals by cross-linked binding proteins
AU2009298879A1 (en) 2008-09-23 2010-04-08 President And Fellows Of Harvard College SIRT4 and uses thereof
WO2010036856A2 (en) 2008-09-26 2010-04-01 Wyeth Llc Compatible display vector systems
KR102097887B1 (en) 2008-09-26 2020-04-06 다나-파버 캔서 인스티튜트 인크. Human anti-pd-1, pd-l1, and pd-l2 antibodies and uses therefor
SI2340038T1 (en) 2008-10-10 2018-05-31 Children's Medical Center Corporation Biochemically stabilized hiv-1 env trimer vaccine
JP2012505900A (en) 2008-10-14 2012-03-08 ダイアクス コーポレーション Use of an IGF-II / IGF-IIE binding protein for the treatment and prevention of pulmonary fibrosis associated with systemic scleroderma
AU2009347206C1 (en) 2008-10-20 2016-12-08 Abbvie Inc. Isolation and purification of antibodies using Protein A affinity chromatography
US10118962B2 (en) 2008-10-29 2018-11-06 Ablynx N.V. Methods for purification of single domain antigen binding molecules
CN104740631B (en) 2008-10-29 2019-04-16 阿布林克斯公司 The preparation of single domain antigen-binding molecule
ES2631507T3 (en) 2008-10-29 2017-08-31 China Synthetic Rubber Corporation Procedures and agents for the diagnosis and treatment of hepatocellular carcinoma
KR20110094029A (en) 2008-11-10 2011-08-19 알렉시온 파마슈티칼스, 인코포레이티드 Methods and compositions for treating complement-associated disorders
AU2009319772A1 (en) 2008-11-26 2010-06-03 Centocor Research & Development, Inc. Compositions and methods for regulating collagen and smooth muscle actin expression by SERPINE2
WO2010063011A2 (en) 2008-11-28 2010-06-03 Emory University Methods for the treatment of infections and tumors
MX2011006483A (en) 2008-12-16 2011-07-13 Novartis Ag Yeast display systems.
WO2010080985A1 (en) 2009-01-08 2010-07-15 Dana-Farber Cancer Institute, Inc. Compositions and methods for induced brown fat differentiation
CA2749966A1 (en) 2009-01-29 2010-08-05 Abbott Laboratories Il-1 binding proteins
US8241623B1 (en) 2009-02-09 2012-08-14 David Bermudes Protease sensitivity expression system
US8030026B2 (en) 2009-02-24 2011-10-04 Abbott Laboratories Antibodies to troponin I and methods of use thereof
WO2010102167A1 (en) 2009-03-05 2010-09-10 Becton, Dickinson And Company Matrix metalloproteinase-7 (mmp-7) monoclonal antibodies and methods for their use in the detection of ovarian cancer
JP5836807B2 (en) 2009-03-05 2015-12-24 アッヴィ・インコーポレイテッド IL-17 binding protein
EP2403875A1 (en) 2009-03-06 2012-01-11 Tripath Imaging, Inc. Glycodelin monoclonal antibodies and methods for their use in the detection of ovarian cancer
WO2010104114A1 (en) * 2009-03-10 2010-09-16 独立行政法人産業技術総合研究所 Method for preparing polypeptide library
EP2414543B1 (en) 2009-04-03 2016-10-12 Decode Genetics EHF Genetic markers for risk management of atrial fibrillation and stroke
EP2424891B1 (en) 2009-04-29 2014-06-11 The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc. Erg monoclonal antibodies
BRPI1012162A2 (en) 2009-04-29 2016-01-12 Abbott Biotech Ltd automatic injection device
BRPI1011195B1 (en) 2009-05-20 2020-10-13 Novimmune S.A methods to produce a collection of nucleic acids
RU2569187C2 (en) 2009-05-29 2015-11-20 МорфоСис АГ Collection and methods of its application
EP2261242A1 (en) 2009-06-10 2010-12-15 Universite Catholique De Louvain Aspartate-N-acetyltransferase enzyme, diagnostic method and therapeutic method
GB0910725D0 (en) 2009-06-22 2009-08-05 Heptares Therapeutics Ltd Mutant proteins and methods for producing them
EP2272979A1 (en) 2009-06-30 2011-01-12 Centre National de la Recherche Scientifique (CNRS) Method for testing a subject thought to be predisposed to having cancer
CN102481380A (en) 2009-07-09 2012-05-30 霍夫曼-拉罗奇有限公司 In vivo tumor vasculature imaging
AU2010269841A1 (en) 2009-07-10 2012-02-23 Decode Genetics Ehf Genetic markers associated with risk of diabetes mellitus
US9259476B2 (en) 2009-07-31 2016-02-16 Wayne State University Monophosphorylated lipid A derivatives
CN102596220A (en) 2009-07-31 2012-07-18 韦恩州立大学 Monophosphorylated lipid A derivatives
CN105131112A (en) 2009-08-29 2015-12-09 Abbvie公司 Therapeutic dll4 binding proteins
EP2473524A4 (en) 2009-09-01 2013-05-22 Abbott Lab Dual variable domain immunoglobulins and uses thereof
US20110189183A1 (en) 2009-09-18 2011-08-04 Robert Anthony Williamson Antibodies against candida, collections thereof and methods of use
US8926976B2 (en) 2009-09-25 2015-01-06 Xoma Technology Ltd. Modulators
CN103403026B (en) 2009-09-25 2016-05-11 美国政府(由卫生和人类服务部的部长所代表) HIV-1 neutralizing antibody and uses thereof
WO2011038301A2 (en) 2009-09-25 2011-03-31 Xoma Technology Ltd. Screening methods
EP2483407A2 (en) 2009-09-30 2012-08-08 President and Fellows of Harvard College Methods for modulation of autophagy through the modulation of autophagy-enhancing gene products
EP2488658A4 (en) 2009-10-15 2013-06-19 Abbvie Inc Dual variable domain immunoglobulins and uses thereof
EP2491055A2 (en) 2009-10-20 2012-08-29 Abbott Laboratories Isolation and purification of anti-il-13 antibodies using protein a affinity chromatography
US20110098862A1 (en) 2009-10-27 2011-04-28 ExxonMobil Research Engineering Company Law Department Multi-stage processes and control thereof
UY32979A (en) 2009-10-28 2011-02-28 Abbott Lab IMMUNOGLOBULINS WITH DUAL VARIABLE DOMAIN AND USES OF THE SAME
WO2011053707A1 (en) 2009-10-31 2011-05-05 Abbott Laboratories Antibodies to receptor for advanced glycation end products (rage) and uses thereof
WO2011058087A1 (en) 2009-11-11 2011-05-19 Gentian As Immunoassay for assessing related analytes of different origin
US8968740B2 (en) 2009-11-13 2015-03-03 Dana-Farber Cancer Institute, Inc. Compositions, kits, and methods for the diagnosis, prognosis, monitoring, treatment and modulation of post-transplant lymphoproliferative disorders and hypoxia associated angiogenesis disorders using galectin-1
MX2012006560A (en) 2009-12-08 2012-10-05 Abbott Gmbh & Co Kg Monoclonal antibodies against the rgm a protein for use in the treatment of retinal nerve fiber layer degeneration.
JP5972793B2 (en) 2009-12-23 2016-08-17 アフィニティ バイオサイエンス ピーティーワイ リミテッド Protein display
CA3168591A1 (en) 2010-01-06 2011-07-14 Takeda Pharmaceutical Company Limited Plasma kallikrein binding proteins
CN102906115A (en) 2010-01-11 2013-01-30 阿雷克森制药公司 Biomarkers of immunomodulatory effects in humans treated with anti-cd200 antibodies
US9745589B2 (en) 2010-01-14 2017-08-29 Cornell University Methods for modulating skeletal remodeling and patterning by modulating SHN2 activity, SHN3 activity, or SHN2 and SHN3 activity in combination
JP2013520162A (en) 2010-01-22 2013-06-06 デイナ ファーバー キャンサー インスティチュート,インコーポレイテッド Compositions, kits and methods for identification, assessment, prevention and treatment of metabolic disorders
WO2011097301A2 (en) 2010-02-02 2011-08-11 Abbott Biotechnology Ltd. METHODS AND COMPOSITIONS FOR PREDICTING RESPONSIVENESS TO TREATMENT WITH TNF-α INHIBITOR
TWI518325B (en) 2010-02-04 2016-01-21 自治醫科大學 Identification, assessment, and therapy of cancers with innate or acquired resistance to alk inhibitors
US8524220B1 (en) 2010-02-09 2013-09-03 David Gordon Bermudes Protease inhibitor: protease sensitivity expression system composition and methods improving the therapeutic activity and specificity of proteins delivered by bacteria
US9597379B1 (en) 2010-02-09 2017-03-21 David Gordon Bermudes Protease inhibitor combination with therapeutic proteins including antibodies
US8771669B1 (en) 2010-02-09 2014-07-08 David Gordon Bermudes Immunization and/or treatment of parasites and infectious agents by live bacteria
MY160628A (en) 2010-03-02 2017-03-15 Abbvie Inc Therapeutic DLL4 Binding Proteins
WO2011119484A1 (en) 2010-03-23 2011-09-29 Iogenetics, Llc Bioinformatic processes for determination of peptide binding
JP2013523182A (en) 2010-04-15 2013-06-17 アボット・ラボラトリーズ Amyloid beta-binding protein
PE20130205A1 (en) 2010-05-14 2013-03-24 Abbvie Inc IL-1 BINDING PROTEINS
CN105056232A (en) 2010-06-03 2015-11-18 阿布维生物技术有限公司 Uses and compositions for treatment of hidradenitis suppurativa
US9405884B2 (en) 2010-06-16 2016-08-02 Abbvie Inc. Methods and systems for the analysis of protein samples
SG186302A1 (en) 2010-06-16 2013-02-28 Abbott Lab Comparison of protein samples
WO2012006500A2 (en) 2010-07-08 2012-01-12 Abbott Laboratories Monoclonal antibodies against hepatitis c virus core protein
UY33492A (en) 2010-07-09 2012-01-31 Abbott Lab IMMUNOGLOBULINS WITH DUAL VARIABLE DOMAIN AND USES OF THE SAME
CN103119062A (en) 2010-07-16 2013-05-22 埃博灵克斯股份有限公司 Modified single domain antigen binding molecules and uses thereof
DK2601609T3 (en) 2010-08-02 2017-06-06 Population Bio Inc COMPOSITIONS AND METHODS FOR DISCOVERING MUTATIONS CAUSING GENETIC DISORDERS
EP3252072A3 (en) 2010-08-03 2018-03-14 AbbVie Inc. Dual variable domain immunoglobulins and uses thereof
WO2012019061A2 (en) 2010-08-05 2012-02-09 Stem Centrx, Inc. Novel effectors and methods of use
EP2420250A1 (en) 2010-08-13 2012-02-22 Universitätsklinikum Münster Anti-Syndecan-4 antibodies
EP2603524A1 (en) 2010-08-14 2013-06-19 AbbVie Inc. Amyloid-beta binding proteins
EP2606067B1 (en) 2010-08-19 2018-02-21 Zoetis Belgium S.A. Anti-ngf antibodies and their use
KR20130139884A (en) 2010-08-26 2013-12-23 애브비 인코포레이티드 Dual variable domain immunoglobulins and uses thereof
EP2608807A1 (en) 2010-08-27 2013-07-03 Stem Centrx, Inc. Notum protein modulators and methods of use
EP2611464B1 (en) 2010-09-03 2018-04-25 AbbVie Stemcentrx LLC Novel modulators and methods of use
CA2810909A1 (en) 2010-09-20 2012-03-29 Abbvie Inc. Purification of antibodies using simulated moving bed chromatography
WO2012044921A1 (en) 2010-10-01 2012-04-05 St. Jude Children's Research Hospital Methods and compositions for typing molecular subgroups of medulloblastoma
WO2012052391A1 (en) 2010-10-19 2012-04-26 Glaxo Group Limited Polypeptide with jmjd3 catalytic activity
US9072766B2 (en) 2010-11-18 2015-07-07 Beth Israel Deaconess Medical Center, Inc. Methods of treating obesity by inhibiting nicotinamide N-methyl transferase (NNMT)
RU2603080C2 (en) 2010-11-19 2016-11-20 МорфоСис АГ Collection and methods for its use
SG190990A1 (en) 2010-12-08 2013-07-31 Stem Centrx Inc Novel modulators and methods of use
SG191312A1 (en) 2010-12-21 2013-07-31 Abbvie Inc Il-1 -alpha and -beta bispecific dual variable domain immunoglobulins and their use
US20120275996A1 (en) 2010-12-21 2012-11-01 Abbott Laboratories IL-1 Binding Proteins
US9394537B2 (en) 2010-12-22 2016-07-19 President And Fellows Of Harvard College Continuous directed evolution
US20140038842A1 (en) 2010-12-28 2014-02-06 Xoma Technology Cell surface display using pdz domains
EP2658869B1 (en) 2010-12-30 2019-06-12 INSERM (Institut National de la Santé et de la Recherche Médicale) Antigen binding formats for use in therapeutic treatments or diagnostic assays
PE20141436A1 (en) 2011-01-24 2014-11-15 Abbvie Biotechnology Ltd AUTOMATIC INJECTION DEVICES WITH OVERMOLDED GRIPPING SURFACES
AU2012212066A1 (en) 2011-02-03 2013-08-15 Alexion Pharmaceuticals, Inc. Use of an anti-CD200 antibody for prolonging the survival of allografts
WO2012109238A2 (en) 2011-02-07 2012-08-16 President And Fellows Of Harvard College Methods for increasing immune responses using agents that directly bind to and activate ire-1
SA112330278B1 (en) 2011-02-18 2015-10-09 ستيم سينتركس، انك. Novel modulators and methods of use
ES2777894T3 (en) 2011-03-02 2020-08-06 Berg Llc Cell-based interrogation assays and uses thereof
AU2012225574A1 (en) 2011-03-07 2013-09-26 University Of Louisville Research Foundation, Inc. Predictive marker of DNMT1 inhibitor therapeutic efficacy and methods of using the marker
US20120238730A1 (en) 2011-03-15 2012-09-20 Abbott Laboratories Integrated approach to the isolation and purification of antibodies
AU2012237287B2 (en) 2011-03-30 2016-09-08 Ablynx Nv Methods of treating immune disorders with single domain antibodies against TNF-alpha
US9777332B2 (en) 2011-03-31 2017-10-03 St. Jude Children's Research Hospital Methods and compositions for identifying minimal residual disease in acute lymphoblastic leukemia
CA2831247C (en) 2011-03-31 2020-07-21 Oriental Yeast Co., Ltd. Cancer immunopotentiating agent containing rankl antagonist
US20120328567A1 (en) 2011-04-08 2012-12-27 Steven Bushnell Biomarkers predictive of therapeutic responsiveness to ifnb and uses thereof
US9150644B2 (en) 2011-04-12 2015-10-06 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Human monoclonal antibodies that bind insulin-like growth factor (IGF) I and II
MX343659B (en) 2011-06-02 2016-11-16 Dyax Corp Fc RECEPTOR BINDING PROTEINS.
ES2894398T3 (en) 2011-06-03 2022-02-14 Xoma Technology Ltd Specific antibodies to TGF-beta
EP2726652B1 (en) 2011-06-29 2016-09-28 Affinity Biosciences Pty Ltd Method of protein display
WO2013009521A2 (en) 2011-07-13 2013-01-17 Abbvie Inc. Methods and compositions for treating asthma using anti-il-13 antibodies
CA2846197C (en) 2011-08-23 2024-01-16 Foundation Medicine, Inc. Kif5b-ret fusion molecules and uses thereof
US20130058947A1 (en) 2011-09-02 2013-03-07 Stem Centrx, Inc Novel Modulators and Methods of Use
US8969519B2 (en) 2011-09-13 2015-03-03 Dana-Farber Cancer Institute, Inc. Compositions and methods for brown fat induction and activity using FNDC5
EP2771349B1 (en) 2011-09-16 2020-02-26 Iogenetics, LLC. Bioinformatic processes for determination of peptide binding
EP2766483B1 (en) 2011-10-10 2022-03-23 The Hospital For Sick Children Methods and compositions for screening and treating developmental disorders
CA2853357A1 (en) 2011-10-24 2013-05-02 Abbvie Inc. Immunobinders directed against tnf
UY34411A (en) 2011-10-24 2013-05-31 Abbvie Inc IMMUNO LINKERS AGAINST SCLEROSTINE
JP6301258B2 (en) 2011-11-03 2018-03-28 トライパス イメージング インコーポレイテッド Methods and compositions for preparing immunostained samples
DK2773779T3 (en) 2011-11-04 2020-11-23 Population Bio Inc METHODS AND COMPOSITIONS FOR DIAGNOSIS, FORECAST AND PREVENTION OF NEUROLOGICAL CONDITIONS
IN2014CN04183A (en) 2011-11-08 2015-07-17 Pfizer
WO2013080050A2 (en) 2011-11-30 2013-06-06 Universitaetsklinikum Erlangen Methods and compositions for determining responsiveness to treatment with a tnf-alpha inhibitor
AU2012352168C1 (en) 2011-12-14 2018-01-25 AbbVie Deutschland GmbH & Co. KG Composition and method for the diagnosis and treatment of iron-related disorders
CA2855570A1 (en) 2011-12-14 2013-06-20 AbbVie Deutschland GmbH & Co. KG Composition and method for the diagnosis and treatment of iron-related disorders
CN104144946A (en) 2011-12-19 2014-11-12 爱克索马美国有限责任公司 Methods for treating acne
CN104159920A (en) 2011-12-30 2014-11-19 艾伯维公司 Dual specific binding proteins directed against il-13 and/or il-17
RS57603B1 (en) 2012-01-27 2018-11-30 Abbvie Deutschland Composition and method for diagnosis and treatment of diseases associated with neurite degeneration
US10407724B2 (en) 2012-02-09 2019-09-10 The Hospital For Sick Children Methods and compositions for screening and treating developmental disorders
ES2812849T3 (en) 2012-02-24 2021-03-18 Abbvie Stemcentrx Llc Anti-DLL3 antibodies and procedures for using them
MX357392B (en) 2012-04-02 2018-07-06 Berg Llc Interrogatory cell-based assays and uses thereof.
SG10201607371UA (en) 2012-04-20 2016-10-28 Merus Nv Methods and means for the production of ig-like molecules
US9181572B2 (en) 2012-04-20 2015-11-10 Abbvie, Inc. Methods to modulate lysine variant distribution
WO2013158279A1 (en) 2012-04-20 2013-10-24 Abbvie Inc. Protein purification methods to reduce acidic species
WO2013158275A1 (en) 2012-04-20 2013-10-24 Abbvie Inc. Cell culture methods to reduce acidic species
US9169304B2 (en) 2012-05-01 2015-10-27 Pfenex Inc. Process for purifying recombinant Plasmodium falciparum circumsporozoite protein
EP2849787A4 (en) 2012-05-14 2016-06-15 Biogen Ma Inc Lingo-2 antagonists for treatment of conditions involving motor neurons
WO2013177115A2 (en) 2012-05-21 2013-11-28 Abbvie Inc. Novel purification of human, humanized, or chimeric antibodies using protein a affinity chromatography
WO2013176754A1 (en) 2012-05-24 2013-11-28 Abbvie Inc. Novel purification of antibodies using hydrophobic interaction chromatography
WO2013184871A1 (en) 2012-06-06 2013-12-12 Zoetis Llc Caninized anti-ngf antibodies and methods thereof
BR112014032916A2 (en) 2012-06-28 2017-08-01 Pfizer anti-drug antibodies and their uses for drug monitoring
WO2014011955A2 (en) 2012-07-12 2014-01-16 Abbvie, Inc. Il-1 binding proteins
WO2014022759A1 (en) 2012-08-03 2014-02-06 Dana-Farber Cancer Institute, Inc. Agents that modulate immune cell activation and methods of use thereof
WO2014024914A1 (en) * 2012-08-08 2014-02-13 第一三共株式会社 Peptide library and use thereof
US9737493B2 (en) 2012-09-07 2017-08-22 University Of Louisville Research Foundation, Inc. Compositions and methods for modulating DNMT1 inhibitor activity
DK2895621T3 (en) 2012-09-14 2020-11-30 Population Bio Inc METHODS AND COMPOSITION FOR DIAGNOSIS, FORECAST AND TREATMENT OF NEUROLOGICAL CONDITIONS
WO2014052855A1 (en) 2012-09-27 2014-04-03 Population Diagnostics, Inc. Methods and compositions for screening and treating developmental disorders
EA201590709A1 (en) 2012-10-09 2015-09-30 Байоджен Айдек Ма Инк. COMBINED THERAPY AND ITS APPLICATION FOR THE TREATMENT OF DIMIELINIZING DISORDERS
KR20180008921A (en) 2012-11-01 2018-01-24 애브비 인코포레이티드 Anti-vegf/dll4 dual variable domain immunoglobulins and uses thereof
CA2890207A1 (en) 2012-11-05 2014-05-08 Foundation Medicine, Inc. Novel ntrk1 fusion molecules and uses thereof
WO2014074942A1 (en) 2012-11-08 2014-05-15 Illumina, Inc. Risk variants of alzheimer's disease
AU2013342163B2 (en) 2012-11-08 2018-08-16 F. Hoffmann-La Roche Ltd IL-6 antagonists and uses thereof
PT2928923T (en) 2012-12-10 2020-03-27 Biogen Ma Inc Anti-blood dendritic cell antigen 2 antibodies and uses thereof
US10036007B2 (en) * 2012-12-11 2018-07-31 Celemics, Inc. Method of synthesis of gene library using codon randomization and mutagenesis
WO2014100542A1 (en) 2012-12-21 2014-06-26 Abbvie, Inc. High-throughput antibody humanization
US10717965B2 (en) 2013-01-10 2020-07-21 Gloriana Therapeutics, Inc. Mammalian cell culture-produced neublastin antibodies
US10980804B2 (en) 2013-01-18 2021-04-20 Foundation Medicine, Inc. Methods of treating cholangiocarcinoma
US9593339B1 (en) 2013-02-14 2017-03-14 David Gordon Bermudes Bacteria carrying bacteriophage and protease inhibitors for the treatment of disorders and methods of treatment
BR112015019909A2 (en) 2013-02-22 2017-08-29 Abbvie Stemcentrx Llc ANTIBODY-DRUG CONJUGATES, PHARMACEUTICAL COMPOSITION, THEIR USES, AND KIT
WO2014142882A1 (en) 2013-03-14 2014-09-18 Abbvie Inc. Protein purification using displacement chromatography
CA2899449A1 (en) 2013-03-14 2014-10-02 Abbvie Inc. Low acidic species compositions and methods for producing the same using displacement chromatography
CA2906421C (en) 2013-03-14 2022-08-16 George J. Dawson Hcv antigen-antibody combination assay and methods and compositions for use therein
SG11201504260UA (en) 2013-03-14 2015-10-29 Abbvie Inc Low acidic species compositions and methods for producing and using the same
SG11201507563SA (en) 2013-03-14 2015-10-29 Parkash Gill Cancer treatment using antibodies that bind cell surface grp78
MX2015012825A (en) 2013-03-14 2016-06-10 Abbott Lab Hcv core lipid binding domain monoclonal antibodies.
BR112015023355A8 (en) 2013-03-14 2018-01-30 Abbott Lab hcv ns3 recombinant antigens and mutants thereof for enhanced antibody detection.
WO2014144292A2 (en) 2013-03-15 2014-09-18 Sanofi Pasteur Biologics , Llc Antibodies against clostridium difficile toxins and methods of using the same
US9469686B2 (en) 2013-03-15 2016-10-18 Abbott Laboratories Anti-GP73 monoclonal antibodies and methods of obtaining the same
EA201591806A1 (en) 2013-03-15 2016-01-29 Байоджен Ма Инк. TREATMENT AND PREVENTION OF ACUTE RENAL FAILURE WITH THE USE OF ANTI-ALPHA-V-BETA-5 ANTIBODIES
CN105324396A (en) 2013-03-15 2016-02-10 艾伯维公司 Dual specific binding proteins directed against il-1 beta and il-17
WO2014168933A1 (en) 2013-04-08 2014-10-16 Cytodyn Inc. Felinized antibodies and methods of treating retroviral infections in felines
WO2014197369A1 (en) 2013-06-06 2014-12-11 Dana-Farber Cancer Institute, Inc. Compositions and methods for identification, assessment prevention, and treatment of cancer using pd-l1 isoforms
EP3632467B1 (en) 2013-06-07 2023-09-27 Duke University Inhibitors of complement factor h
WO2015017529A2 (en) 2013-07-31 2015-02-05 Dana-Farber Cancer Institute, Inc. Compositions and methods for modulating thermogenesis using pth-related and egf-related molecules
EP3036320B2 (en) 2013-08-19 2024-04-03 Biogen MA Inc. Control of protein glycosylation by culture medium supplementation and cell culture process parameters
AU2014312215B2 (en) 2013-08-28 2020-02-27 Abbvie Stemcentrx Llc Site-specific antibody conjugation methods and compositions
CN105792836A (en) 2013-08-28 2016-07-20 施特姆森特克斯股份有限公司 Novel SEZ6 modulators and methods of use
EP3521431A1 (en) 2013-09-25 2019-08-07 Cornell University Compounds for inducing anti-tumor immunity and methods thereof
ES2900425T3 (en) 2013-09-25 2022-03-16 Bioverativ Therapeutics Inc Column viral inactivation methods
EP3757130A1 (en) 2013-09-26 2020-12-30 Costim Pharmaceuticals Inc. Methods for treating hematologic cancers
WO2015057939A1 (en) 2013-10-18 2015-04-23 Biogen Idec Ma Inc. Anti-s1p4 antibodies and uses thereof
WO2015066190A1 (en) 2013-10-29 2015-05-07 President And Fellows Of Harvard College Methods and compositions for inhibting oxidative stress
EP3066121A1 (en) 2013-11-07 2016-09-14 AbbVie Inc. Isolation and purification of dvd-igs
EP2891722B1 (en) 2013-11-12 2018-10-10 Population Bio, Inc. Methods and compositions for diagnosing, prognosing, and treating endometriosis
CA2928014A1 (en) 2013-11-15 2015-05-21 Institut Pasteur Method for detecting a molecular marker of plasmodium falciparum artemisinin resistance
CA2931684C (en) 2013-12-19 2024-02-20 Novartis Ag Human mesothelin chimeric antigen receptors and uses thereof
WO2015095809A1 (en) 2013-12-20 2015-06-25 Biogen Idec Ma Inc. Use of perfusion seed cultures to improve biopharmaceutical fed-batch production capacity and product quality
US11708411B2 (en) 2013-12-20 2023-07-25 Wake Forest University Health Sciences Methods and compositions for increasing protective antibody levels induced by pneumococcal polysaccharide vaccines
EP2960252A1 (en) 2014-06-26 2015-12-30 Institut Pasteur Phospholipase for treatment of immunosuppression
EP3097196B1 (en) 2014-01-20 2019-09-11 President and Fellows of Harvard College Negative selection and stringency modulation in continuous evolution systems
JOP20200094A1 (en) 2014-01-24 2017-06-16 Dana Farber Cancer Inst Inc Antibody molecules to pd-1 and uses thereof
JOP20200096A1 (en) 2014-01-31 2017-06-16 Children’S Medical Center Corp Antibody molecules to tim-3 and uses thereof
CN106211773B (en) 2014-02-11 2021-09-03 威特拉公司 Antibody molecules for dengue virus and uses thereof
KR20170008202A (en) 2014-02-21 2017-01-23 애브비 스템센트알엑스 엘엘씨 Anti-dll3 antibodies and drug conjugates for use in melanoma
PT3116909T (en) 2014-03-14 2020-01-30 Novartis Ag Antibody molecules to lag-3 and uses thereof
EP3122869B2 (en) 2014-03-24 2022-08-10 Biogen MA Inc. Methods for overcoming glutamine deprivation during mammalian cell culture
AU2015259516B2 (en) 2014-05-13 2020-05-28 Bavarian Nordic A/S Combination therapy for treating cancer with a poxvirus expressing a tumor antigen and a monoclonal antibody against TIM-3
MX355107B (en) 2014-06-03 2018-04-04 Xbiotech Inc Compositions and methods for treating and preventing staphylococcus aureus infections.
US10626375B2 (en) 2014-06-25 2020-04-21 Auxergen, Inc. Disease control of the plant bacterial pathogens causing citrus canker and rice blight
KR102594343B1 (en) 2014-07-21 2023-10-26 노파르티스 아게 Treatment of cancer using a cd33 chimeric antigen receptor
EP3172228B1 (en) 2014-07-25 2019-02-27 Theravectys Lentiviral vectors for regulated expression of a chimeric antigen receptor molecule
CA2958200A1 (en) 2014-08-14 2016-02-18 Novartis Ag Treatment of cancer using a gfr alpha-4 chimeric antigen receptor
PT3183268T (en) 2014-08-19 2020-05-15 Novartis Ag Anti-cd123 chimeric antigen receptor (car) for use in cancer treatment
US10724096B2 (en) 2014-09-05 2020-07-28 Population Bio, Inc. Methods and compositions for inhibiting and treating neurological conditions
EP3191097B1 (en) 2014-09-13 2019-10-23 Novartis AG Combination therapies
EP3194444B1 (en) 2014-09-16 2019-07-10 Symphogen A/S Anti-met antibodies and compositions
RU2754684C2 (en) 2014-09-30 2021-09-06 Дойчес Кребсфоршунгсцентрум Штифтунг Дес Эффентлихен Рехтс Binding molecules, namely antibodies capable of binding to l1cam (cd171)
KR20170066546A (en) 2014-10-03 2017-06-14 노파르티스 아게 Combination therapies
MX2017004810A (en) 2014-10-14 2017-10-16 Novartis Ag Antibody molecules to pd-l1 and uses thereof.
MA41685A (en) 2014-10-17 2017-08-22 Biogen Ma Inc COPPER SUPPLEMENT FOR THE REGULATION OF GLYCOSYLATION IN A MAMMAL CELL CULTURE PROCESS
US10920208B2 (en) 2014-10-22 2021-02-16 President And Fellows Of Harvard College Evolution of proteases
MA40864A (en) 2014-10-31 2017-09-05 Biogen Ma Inc HYPOTAURINE, GABA, BETA-ALANINE AND CHOLINE FOR THE REGULATION OF THE ACCUMULATION OF RESIDUAL BY-PRODUCTS IN MAMMAL CELL CULTURE PROCESSES
SG11201703667SA (en) 2014-11-05 2017-06-29 Annexon Inc Humanized anti-complement factor c1q antibodies and uses thereof
WO2016073894A1 (en) 2014-11-07 2016-05-12 Eleven Biotherapeutics, Inc. Therapeutic agents with increased ocular retention
UA122673C2 (en) 2014-11-07 2020-12-28 Елевен Байотерапьютікс, Інк. Improved il-6 antibodies
EP3220900B1 (en) 2014-11-21 2020-09-23 University of Maryland, Baltimore Targeted structure-specific particulate delivery systems
US11220545B2 (en) 2014-12-08 2022-01-11 Dana-Farber Cancer Institute, Inc. Methods for upregulating immune responses using combinations of anti-RGMb and anti-PD-1 agents
US10093733B2 (en) 2014-12-11 2018-10-09 Abbvie Inc. LRP-8 binding dual variable domain immunoglobulin proteins
WO2016100882A1 (en) 2014-12-19 2016-06-23 Novartis Ag Combination therapies
JP2018504400A (en) 2015-01-08 2018-02-15 バイオジェン・エムエイ・インコーポレイテッドBiogen MA Inc. LINGO-1 antagonist and use for treatment of demyelinating disorders
US10711067B2 (en) 2015-03-03 2020-07-14 Xoma (Us) Llc Treatment of post-prandial hyperinsulinemia and hypoglycemia after bariatric surgery
EP3735982A1 (en) 2015-03-10 2020-11-11 The University of Massachusetts Targeting gdf6 and bmp signaling for anti-melanoma therapy
JP6901400B2 (en) 2015-04-03 2021-07-14 ゾーマ テクノロジー リミテッド Cancer treatment using TGF-β and PD-1 inhibitors
WO2016168631A1 (en) 2015-04-17 2016-10-20 President And Fellows Of Harvard College Vector-based mutagenesis system
EP3285811A1 (en) 2015-04-21 2018-02-28 Institut Gustave Roussy Therapeutic methods, products and compositions inhibiting znf555
WO2016183183A1 (en) 2015-05-11 2016-11-17 The Johns Hopkins University Autoimmune antibodies for use in inhibiting cancer cell growth
JP6518917B2 (en) 2015-05-29 2019-05-29 アッヴィ・インコーポレイテッド Anti-CD40 antibody and use thereof
TW201710286A (en) 2015-06-15 2017-03-16 艾伯維有限公司 Binding proteins against VEGF, PDGF, and/or their receptors
WO2017015545A1 (en) 2015-07-22 2017-01-26 President And Fellows Of Harvard College Evolution of site-specific recombinases
WO2017015559A2 (en) 2015-07-23 2017-01-26 President And Fellows Of Harvard College Evolution of bt toxins
DK3317301T3 (en) 2015-07-29 2021-06-28 Immutep Sas COMBINATION THERAPIES INCLUDING ANTIBODY MOLECULES AGAINST LAYER-3
US20180207273A1 (en) 2015-07-29 2018-07-26 Novartis Ag Combination therapies comprising antibody molecules to tim-3
US10612011B2 (en) 2015-07-30 2020-04-07 President And Fellows Of Harvard College Evolution of TALENs
JP2018532693A (en) 2015-08-06 2018-11-08 ゾーマ (ユーエス) リミテッド ライアビリティ カンパニー Antibody fragments against the insulin receptor and their use to treat hypoglycemia
EP3932953A1 (en) 2015-08-28 2022-01-05 F. Hoffmann-La Roche AG Anti-hypusine antibodies and uses thereof
RS62330B1 (en) 2015-09-15 2021-10-29 Scholar Rock Inc Anti-pro/latent-myostatin antibodies and uses thereof
EP3362074B1 (en) 2015-10-16 2023-08-09 President and Fellows of Harvard College Regulatory t cell pd-1 modulation for regulating t cell effector immune responses
AU2016343978A1 (en) 2015-10-29 2018-05-17 Dana-Farber Cancer Institute, Inc. Methods for identification, assessment, prevention, and treatment of metabolic disorders using PM20D1 and N-lipidated amino acids
JP7041628B2 (en) 2015-11-10 2022-03-24 ビステラ, インコーポレイテッド Antibody molecules that specifically bind to lipopolysaccharide-drug conjugates and their use
MX2018006385A (en) 2015-11-25 2019-09-11 Visterra Inc Antibody molecules to april and uses thereof.
EP3389712B1 (en) 2015-12-17 2024-04-10 Novartis AG Antibody molecules to pd-1 and uses thereof
US20200261573A1 (en) 2015-12-17 2020-08-20 Novartis Ag Combination of c-met inhibitor with antibody molecule to pd-1 and uses thereof
AU2017206069A1 (en) 2016-01-08 2018-07-19 Scholar Rock, Inc. Anti-pro/latent myostatin antibodies and methods of use thereof
CA3015913A1 (en) 2016-02-29 2017-09-08 Foundation Medicine, Inc. Methods of treating cancer
EP4169942A1 (en) 2016-03-11 2023-04-26 Scholar Rock, Inc. Tgfbeta1-binding immunoglobulins and use thereof
WO2017160599A1 (en) 2016-03-14 2017-09-21 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Use of cd300b antagonists to treat sepsis and septic shock
WO2017158436A1 (en) 2016-03-17 2017-09-21 Oslo Universitetssykehus Hf Fusion proteins targeting tumour associated macrophages for treating cancer
CA3016563A1 (en) 2016-03-21 2017-09-28 Elstar Therapeutics, Inc. Multispecific and multifunctional molecules and uses thereof
TWI763660B (en) 2016-03-25 2022-05-11 美商威特拉公司 Formulations of antibody molecules to dengue virus
CN116333140A (en) 2016-04-08 2023-06-27 埃缇健康公司D/B/A泽尔拜尔 Reticulin-1 binding antibodies and uses thereof
US20170298119A1 (en) 2016-04-15 2017-10-19 Visterra, Inc. Antibody molecules to zika virus and uses thereof
MA45491A (en) 2016-06-27 2019-05-01 Juno Therapeutics Inc CMH-E RESTRICTED EPITOPES, BINDING MOLECULES AND RELATED METHODS AND USES
CN110291402B (en) 2016-06-27 2023-09-01 朱诺治疗学股份有限公司 Method for identifying peptide epitopes, molecules binding such epitopes and related uses
CA3030745A1 (en) 2016-07-13 2018-01-18 Biogen Ma Inc. Dosage regimens of lingo-1 antagonists and uses for treatment of demyelinating disorders
WO2018013918A2 (en) 2016-07-15 2018-01-18 Novartis Ag Treatment and prevention of cytokine release syndrome using a chimeric antigen receptor in combination with a kinase inhibitor
WO2018022479A1 (en) 2016-07-25 2018-02-01 Biogen Ma Inc. Anti-hspa5 (grp78) antibodies and uses thereof
CN109803680A (en) 2016-08-01 2019-05-24 佐马美国有限公司 (PTH1R) antibody of parathyroid hormone receptor 1 and its purposes
SG11201900616UA (en) 2016-08-02 2019-02-27 Visterra Inc Engineered polypeptides and uses thereof
WO2018027042A1 (en) 2016-08-03 2018-02-08 Bio-Techne Corporation Identification of vsig3/vista as a novel immune checkpoint and use thereof for immunotherapy
WO2018057955A1 (en) 2016-09-23 2018-03-29 Elstar Therapeutics, Inc. Multispecific antibody molecules comprising lambda and kappa light chains
US10858428B2 (en) 2016-09-28 2020-12-08 Xoma (Us) Llc Antibodies that bind interleukin-2 and uses thereof
JP2019535015A (en) 2016-10-03 2019-12-05 アボット・ラボラトリーズAbbott Laboratories Improved method for assessing GFAP status in patient samples
CN110225927B (en) 2016-10-07 2024-01-12 诺华股份有限公司 Chimeric antigen receptor for the treatment of cancer
WO2018071576A1 (en) 2016-10-14 2018-04-19 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Treatment of tumors by inhibition of cd300f
WO2018075408A1 (en) 2016-10-17 2018-04-26 Alexion Pharmaceuticals, Inc. Methods of treating acute myeloid leukemia (aml) with combinations of anti-cd200 antibodies, cytarabine, and daunorubicin
WO2018102594A1 (en) 2016-12-01 2018-06-07 Alexion Pharmaceuticals, Inc. Methods of treating solid tumors with anti-cd200 antibodies
US11180535B1 (en) 2016-12-07 2021-11-23 David Gordon Bermudes Saccharide binding, tumor penetration, and cytotoxic antitumor chimeric peptides from therapeutic bacteria
US11129906B1 (en) 2016-12-07 2021-09-28 David Gordon Bermudes Chimeric protein toxins for expression by therapeutic bacteria
MX2019007356A (en) 2016-12-23 2019-09-05 Visterra Inc Binding polypeptides and methods of making the same.
PT3565592T (en) 2017-01-06 2023-05-31 Scholar Rock Inc Methods for treating metabolic diseases by inhibiting myostatin activation
AU2018205231A1 (en) 2017-01-06 2019-07-18 Scholar Rock, Inc. Isoform-specific, context-permissive TGFβ1 inhibitors and use thereof
HRP20230308T1 (en) 2017-01-06 2023-05-12 Scholar Rock, Inc. Treating metabolic diseases by inhibiting myostatin activation
TWI659751B (en) 2017-01-13 2019-05-21 中央研究院 Improved reloadable hydrogel system for treating brain conditions
WO2018130660A1 (en) 2017-01-13 2018-07-19 Academia Sinica Reloadable hydrogel system for treating myocardial infarction
JP7303110B2 (en) 2017-01-18 2023-07-04 ビステラ, インコーポレイテッド Antibody molecule-drug conjugates and uses thereof
US10240205B2 (en) 2017-02-03 2019-03-26 Population Bio, Inc. Methods for assessing risk of developing a viral disease using a genetic test
AR111211A1 (en) 2017-02-06 2019-06-19 Australian Meat & Live Stock IMMUNO STIMULATING COMPOSITIONS AND THEIR USES
US20200291089A1 (en) 2017-02-16 2020-09-17 Elstar Therapeutics, Inc. Multifunctional molecules comprising a trimeric ligand and uses thereof
US11685776B2 (en) 2017-03-02 2023-06-27 Novartis Ag Engineered heterodimeric proteins
EP3595710A4 (en) * 2017-03-13 2021-01-13 Duke University Antigen display system and methods for characterizing antibody responses
JP7346300B2 (en) 2017-03-23 2023-09-19 アボット・ラボラトリーズ Methods for aiding in the diagnosis and determination of the extent of traumatic brain injury in human subjects using the early biomarker ubiquitin carboxy-terminal hydrolase L1
EP3599843A4 (en) 2017-03-24 2021-01-13 The Regents of the University of California Proteoglycan irregularities in abnormal fibroblasts and therapies based therefrom
AU2018249493A1 (en) 2017-04-03 2019-09-19 Oncxerna Therapeutics, Inc. Methods for treating cancer using PS-targeting antibodies with immuno-oncology agents
JP2020512825A (en) 2017-04-12 2020-04-30 ファイザー・インク Antibodies with conditional affinity and methods of use thereof
US10877048B2 (en) 2017-04-15 2020-12-29 Abbott Laboratories Methods for aiding in the hyperacute diagnosis and determination of traumatic brain injury in a human subject using early biomarkers
JP2020517256A (en) 2017-04-19 2020-06-18 エルスター セラピューティクス, インコーポレイテッド Multispecific molecules and uses thereof
WO2018200823A1 (en) 2017-04-28 2018-11-01 Abbott Laboratories Methods for aiding in the hyperacute diagnosis and determination of traumatic brain injury using early biomarkers on at least two samples from the same human subject
AU2018260545B2 (en) 2017-04-28 2023-11-23 Marengo Therapeutics, Inc. Multispecific molecules comprising a non-immunoglobulin heterodimerization domain and uses thereof
US10865238B1 (en) 2017-05-05 2020-12-15 Duke University Complement factor H antibodies
EP3630830A1 (en) 2017-05-23 2020-04-08 Helmholtz Zentrum München - Deutsches Forschungszentrum für Gesundheit und Umwelt (GmbH) Novel cd73 antibody, preparation and uses thereof
AU2018272054A1 (en) 2017-05-25 2019-09-26 Abbott Laboratories Methods for aiding in the determination of whether to perform imaging on a human subject who has sustained or may have sustained an injury to the head using early biomarkers
BR112019025313A2 (en) 2017-05-30 2020-06-23 Abbott Laboratories METHODS FOR ASSISTANCE IN DIAGNOSIS AND EVALUATION OF A LIGHT TRAUMATIC BRAIN INJURY IN A HUMAN INDIVIDUAL USING HEART TROPONIN I
EP3630836A1 (en) 2017-05-31 2020-04-08 Elstar Therapeutics, Inc. Multispecific molecules that bind to myeloproliferative leukemia (mpl) protein and uses thereof
AU2018281830B2 (en) 2017-06-09 2023-11-02 Agonox, Inc. Utilization of CD39 and CD103 for identification of human tumor reactive cells for treatment of cancer
US20200172628A1 (en) 2017-06-22 2020-06-04 Novartis Ag Antibody molecules to cd73 and uses thereof
EP3645042A4 (en) 2017-06-26 2021-03-17 Bio-Techne Corporation Hybridoma clones, monoclonal antibodies to vsig-4, and methods of making and using
JP2020525483A (en) 2017-06-27 2020-08-27 ノバルティス アーゲー Dosing regimens for anti-TIM-3 antibodies and uses thereof
CA3068041A1 (en) 2017-07-03 2019-01-10 Abbott Laboratories Improved methods for measuring ubiquitin carboxy-terminal hydrolase l1 levels in blood
US11447809B2 (en) 2017-07-06 2022-09-20 President And Fellows Of Harvard College Evolution of tRNA synthetases
SG11202000298VA (en) 2017-07-14 2020-02-27 Pfizer Antibodies to madcam
EP3431496A1 (en) 2017-07-19 2019-01-23 Fraunhofer-Gesellschaft zur Förderung der angewandten Forschung e.V. Anti- isoasp7 amyloid beta antibodies and uses thereof
AU2018302283A1 (en) 2017-07-20 2020-02-06 Novartis Ag Dosage regimens of anti-LAG-3 antibodies and uses thereof
CA3071427A1 (en) 2017-07-28 2019-01-31 Scholar Rock, Inc. Ltbp complex-specific inhibitors of tgf-beta 1 and uses thereof
WO2019035938A1 (en) 2017-08-16 2019-02-21 Elstar Therapeutics, Inc. Multispecific molecules that bind to bcma and uses thereof
WO2019056002A1 (en) 2017-09-18 2019-03-21 President And Fellows Of Harvard College Continuous evolution for stabilized proteins
US11761963B2 (en) 2017-09-27 2023-09-19 Alexion Pharmaceuticals, Inc. Biomarker signature for predicting tumor response to anti-CD200 therapy
CA3074032A1 (en) 2017-10-02 2019-04-11 Visterra, Inc. Antibody molecules to cd138 and uses thereof
ES2759622T3 (en) 2017-10-02 2020-05-11 Certest Biotec S L Anti-Dps antibodies and test devices for the detection of bacteria of the genus Campylobacter
KR20210027230A (en) 2017-10-04 2021-03-10 옵코 파마슈티칼스, 엘엘씨 Articles and methods for personalized treatment of cancer
US20210198365A1 (en) 2017-10-19 2021-07-01 Merck Patent Gmbh Combination product for the treatment of cancer
US20200331975A1 (en) 2017-10-20 2020-10-22 Institut Curie Dap10/12 based cars adapted for rush
MX2020004756A (en) 2017-11-16 2020-08-20 Novartis Ag Combination therapies.
JP7348899B2 (en) 2017-12-08 2023-09-21 マレンゴ・セラピューティクス,インコーポレーテッド Multispecific molecules and their uses
CA3067055A1 (en) 2017-12-09 2019-06-13 Abbott Laboratories Methods for aiding in diagnosing and evaluating a traumatic brain injury in a human subject using a combination of gfap and uch-l1
CN111094983A (en) 2017-12-09 2020-05-01 雅培实验室 Methods of using Glial Fibrillary Acidic Protein (GFAP) and/or ubiquitin carboxy-terminal hydrolase L1(UCH-L1) to aid in the diagnosis and evaluation of patients who have suffered orthopedic injury and who have suffered or may have suffered a head injury such as mild Traumatic Brain Injury (TBI)
US11802154B2 (en) 2017-12-20 2023-10-31 Alexion Pharmaceuticals, Inc. Humanized anti-CD200 antibodies and uses thereof
WO2019126133A1 (en) 2017-12-20 2019-06-27 Alexion Pharmaceuticals, Inc. Liquid formulations of anti-cd200 antibodies
CN111868082A (en) 2018-02-02 2020-10-30 博奥泰克尼公司 Compounds that modulate the interaction of VISTA and VSIG3 and methods of making and using the same
EP3765499A1 (en) 2018-03-12 2021-01-20 Zoetis Services LLC Anti-ngf antibodies and methods thereof
US20210238280A1 (en) 2018-03-14 2021-08-05 Elstar Therapeutics, Inc. Multifunctional molecules that bind to calreticulin and uses thereof
EP3765516A2 (en) 2018-03-14 2021-01-20 Elstar Therapeutics, Inc. Multifunctional molecules and uses thereof
WO2019180272A1 (en) 2018-03-23 2019-09-26 Fundación Instituto De Investigación Sanitaria De Santiago De Compostela Anti-leptin affinity reagents for use in the treatment of obesity and other leptin-resistance associated diseases
CA3091733A1 (en) 2018-03-26 2019-10-03 Glycanostics S.R.O. Means and methods for glycoprofiling of a protein
WO2019190877A1 (en) 2018-03-26 2019-10-03 Alexion Pharmaceuticals, Inc. High throughput method for measuring the protease activity of complement c3 convertase
US20210147547A1 (en) 2018-04-13 2021-05-20 Novartis Ag Dosage Regimens For Anti-Pd-L1 Antibodies And Uses Thereof
TW202015726A (en) 2018-05-30 2020-05-01 瑞士商諾華公司 Entpd2 antibodies, combination therapies, and methods of using the antibodies and combination therapies
US11913044B2 (en) 2018-06-14 2024-02-27 President And Fellows Of Harvard College Evolution of cytidine deaminases
CA3104295A1 (en) 2018-06-19 2019-12-26 Atarga, Llc Antibody molecules to complement component 5 and uses thereof
JP7424998B2 (en) * 2018-06-20 2024-01-30 ホフセス バイオケア アーエスアー Fish protein hydrolyzate powder and compositions comprising said powder for use as a medicine
EP3818083A2 (en) 2018-07-03 2021-05-12 Elstar Therapeutics, Inc. Anti-tcr antibody molecules and uses thereof
WO2020008083A1 (en) 2018-07-05 2020-01-09 Consejo Superior De Investigaciones Científicas Therapeutic target in chemokine receptors for the screening of compounds useful for the treatment of pathological processes involving chemokine signaling
PL3677278T3 (en) 2018-07-11 2022-02-28 Scholar Rock, Inc. Isoform selective tgfbeta1 inhibitors and use thereof
WO2020014473A1 (en) 2018-07-11 2020-01-16 Scholar Rock, Inc. TGFβ1 INHIBITORS AND USE THEREOF
KR20210058811A (en) 2018-07-11 2021-05-24 스칼러 락, 인크. High affinity, isotype-selective TGFβ1 inhibitors and uses thereof
WO2020021465A1 (en) 2018-07-25 2020-01-30 Advanced Accelerator Applications (Italy) S.R.L. Method of treatment of neuroendocrine tumors
EP3625368B8 (en) 2018-08-08 2022-12-14 PML Screening, LLC Methods for assessing the risk of developing progressive multifocal leukoencephalopathy caused by john cunningham virus by genetic testing
US20220242957A1 (en) 2018-09-27 2022-08-04 Marengo Therapeutics, Inc. Csf1r/ccr2 multispecific antibodies
EP3860641A1 (en) 2018-10-05 2021-08-11 Bavarian Nordic A/S Combination therapy for treating cancer with an intravenous administration of a recombinant mva and an immune checkpoint antagonist or agonist
UY38407A (en) 2018-10-15 2020-05-29 Novartis Ag TREM2 STABILIZING ANTIBODIES
SG11202104010PA (en) 2018-10-23 2021-05-28 Scholar Rock Inc Rgmc-selective inhibitors and use thereof
WO2020086408A1 (en) 2018-10-26 2020-04-30 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services A high-yield perfusion-based transient gene expression bioprocess
AU2019385665A1 (en) 2018-11-20 2021-05-27 Bavarian Nordic A/S Therapy for treating cancer with an intratumoral and/or intravenous administration of a recombinant MVA encoding 4-1BBL (CD137L) and/or CD40l
WO2020118011A1 (en) 2018-12-06 2020-06-11 Alexion Pharmaceuticals, Inc. Anti-alk2 antibodies and uses thereof
TW202038959A (en) 2018-12-20 2020-11-01 瑞士商諾華公司 Novel dosing regimens for mdm2 inhibitors
SG11202108099WA (en) 2019-01-30 2021-08-30 Scholar Rock Inc LTBP COMPLEX-SPECIFIC INHIBITORS OF TGFβ AND USES THEREOF
US11738050B2 (en) 2019-02-01 2023-08-29 Regents Of The University Of Minnesota Compounds binding to fibroblast activation protein alpha
EP3693063A1 (en) 2019-02-06 2020-08-12 Diaccurate Methods and compositions for treating cancer
EP3696191A1 (en) 2019-02-14 2020-08-19 Fundación Instituto de Investigación contra la Leucemia Josep Carreras (IJC) Car t-cells for the treatment of cd1a-positive cancer
US10871640B2 (en) 2019-02-15 2020-12-22 Perkinelmer Cellular Technologies Germany Gmbh Methods and systems for automated imaging of three-dimensional objects
GB2599228B (en) 2019-02-21 2024-02-07 Marengo Therapeutics Inc Multifunctional molecules that bind to T cell related cancer cells and uses thereof
SG11202109056TA (en) 2019-02-21 2021-09-29 Marengo Therapeutics Inc Multifunctional molecules that bind to calreticulin and uses thereof
CA3131014A1 (en) 2019-02-21 2020-08-27 Andreas Loew Anti-tcr antibody molecules and uses thereof
CA3130508A1 (en) 2019-02-21 2020-08-27 Marengo Therapeutics, Inc. Antibody molecules that bind to nkp30 and uses thereof
WO2020172598A1 (en) 2019-02-21 2020-08-27 Elstar Therapeutics, Inc. Multifunctional molecules that bind to t cells and uses thereof to treat autoimmune disorders
MA55519A (en) 2019-03-29 2022-02-09 Atarga Llc ANTI-FGF23 ANTIBODIES
KR20220044939A (en) 2019-06-17 2022-04-12 비스테라, 인크. Humanized Antibody Molecules Against CD138 and Uses Thereof
JP2022541657A (en) 2019-07-26 2022-09-26 ビステラ, インコーポレイテッド Interleukin-2 agonists and uses thereof
HRP20240037T1 (en) 2019-08-02 2024-03-29 Fundació de Recerca Clínic Barcelona-Institut d'Investigacions Biomèdiques August Pi i Sunyer (FRCB-IDIBAPS) Car t-cells against bcma for the treatment of multiple myeloma
EP4031658A1 (en) 2019-08-07 2022-07-27 DB Biotech, AS Improved horseradish peroxidase polypeptides
WO2021044009A1 (en) 2019-09-04 2021-03-11 Deutsches Zentrum Für Neurodegenerative Erkrankungen E.V. (Dzne) Herv inhibitors for use in treating tauopathies
US20220348651A1 (en) 2019-09-18 2022-11-03 Novartis Ag Entpd2 antibodies, combination therapies, and methods of using the antibodies and combination therapies
TW202124446A (en) 2019-09-18 2021-07-01 瑞士商諾華公司 Combination therapies with entpd2 antibodies
TW202128191A (en) 2019-10-21 2021-08-01 瑞士商諾華公司 Tim-3 inhibitors and uses thereof
CN114786679A (en) 2019-10-21 2022-07-22 诺华股份有限公司 Combination therapy with Vernetork and TIM-3 inhibitors
BR112022009798A2 (en) 2019-11-20 2022-10-18 Bavarian Nordic As RECOMBINANT MVA VIRUS FOR INTRATUMORAL AND/OR INTRAVENOUS ADMINISTRATION FOR CANCER TREATMENT
WO2021123902A1 (en) 2019-12-20 2021-06-24 Novartis Ag Combination of anti tim-3 antibody mbg453 and anti tgf-beta antibody nis793, with or without decitabine or the anti pd-1 antibody spartalizumab, for treating myelofibrosis and myelodysplastic syndrome
WO2021138407A2 (en) 2020-01-03 2021-07-08 Marengo Therapeutics, Inc. Multifunctional molecules that bind to cd33 and uses thereof
US20230057012A1 (en) 2020-01-11 2023-02-23 Scholar Rock, Inc. Tgfb inhibitors and use thereof
US20230050148A1 (en) 2020-01-11 2023-02-16 Scholar Rock, Inc. Tgf-beta inhibitors and use thereof
TW202140553A (en) 2020-01-13 2021-11-01 美商威特拉公司 Antibody molecules to c5ar1 and uses thereof
IL293752A (en) 2020-01-17 2022-08-01 Novartis Ag Combination comprising a tim-3 inhibitor and a hypomethylating agent for use in treating myelodysplastic syndrome or chronic myelomonocytic leukemia
AU2021215936A1 (en) 2020-02-05 2022-08-25 Larimar Therapeutics, Inc. TAT peptide binding proteins and uses thereof
US20230139885A1 (en) 2020-03-11 2023-05-04 Fundació Institut De Recerca Contra La Leucèmia Josep Carreras Cd22 targeting-moiety for the treatment of b-cell acute lymphoblastic leukemia (b-all)
CN116710146A (en) 2020-04-03 2023-09-05 威特拉公司 Antibody molecule-drug conjugates and uses thereof
EP4136459A1 (en) 2020-04-13 2023-02-22 Abbott Laboratories Methods, complexes and kits for detecting or determining an amount of a ss-coronavirus antibody in a sample
JP2023523011A (en) 2020-04-24 2023-06-01 マレンゴ・セラピューティクス,インコーポレーテッド Multifunctional molecules that bind to T cell-associated cancer cells and uses thereof
CN116096758A (en) 2020-05-01 2023-05-09 诺华股份有限公司 Engineered immunoglobulins
WO2021220218A1 (en) 2020-05-01 2021-11-04 Novartis Ag Immunoglobulin variants
WO2021239666A1 (en) 2020-05-26 2021-12-02 Diaccurate Therapeutic methods
US20230235080A1 (en) 2020-06-03 2023-07-27 Bionecure Therapeutics, Inc. Trophoblast cell-surface antigen-2 (trop-2) antibodies
CA3187823A1 (en) 2020-06-24 2021-12-30 Visterra, Inc. Antibody molecules to april and uses thereof
US20230256114A1 (en) 2020-07-07 2023-08-17 Bionecure Therapeutics, Inc. Novel maytansinoids as adc payloads and their use for the treatment of cancer
WO2022026592A2 (en) 2020-07-28 2022-02-03 Celltas Bio, Inc. Antibody molecules to coronavirus and uses thereof
US20220043000A1 (en) 2020-08-04 2022-02-10 Abbott Laboratories Methods and kits for detecting sars-cov-2 protein in a sample
CN116761818A (en) 2020-08-26 2023-09-15 马伦戈治疗公司 Method for detecting TRBC1 or TRBC2
GB2616128A (en) 2020-08-26 2023-08-30 Marengo Therapeutics Inc Antibody molecules that bind to NKp30 and uses thereof
CA3190755A1 (en) 2020-08-26 2022-03-03 Andreas Loew Multifunctional molecules that bind to calreticulin and uses thereof
EP4204020A1 (en) 2020-08-31 2023-07-05 Advanced Accelerator Applications International S.A. Method of treating psma-expressing cancers
WO2022043557A1 (en) 2020-08-31 2022-03-03 Advanced Accelerator Applications International Sa Method of treating psma-expressing cancers
WO2022081718A1 (en) 2020-10-14 2022-04-21 Five Prime Therapeutics, Inc. Anti-c-c chemokine receptor 8 (ccr8) antibodies and methods of use thereof
US20230408517A1 (en) 2020-11-24 2023-12-21 Bio-Techne Corporation Anti-severe acute respiratory syndrome coronavirus antibodies
WO2023102384A1 (en) 2021-11-30 2023-06-08 Abbott Laboratories Use of one or more biomarkers to determine traumatic brain injury (tbi) in a subject having received a head computerized tomography scan that is negative for a tbi
CA3198161A1 (en) 2020-12-01 2022-06-09 Beth MCQUISTON Use of one or more biomarkers to determine traumatic brain injury (tbi) in a subject having received a head computerized tomography scan that is negative for a tbi
CA3203977A1 (en) 2020-12-04 2022-06-09 Visterra, Inc. Methods of using interleukin-2 agents
CA3202339A1 (en) 2020-12-18 2022-06-23 Mei Jang Protein compositions and methods for producing and using the same
EP4271998A1 (en) 2020-12-30 2023-11-08 Abbott Laboratories Methods for determining sars-cov-2 antigen and anti-sars-cov-2 antibody in a sample
CA3208934A1 (en) 2021-01-20 2022-07-28 Visterra, Inc. Interleukin-2 mutants and uses thereof
EP4284510A1 (en) 2021-01-29 2023-12-06 Novartis AG Dosage regimes for anti-cd73 and anti-entpd2 antibodies and uses thereof
WO2022182872A2 (en) 2021-02-24 2022-09-01 Alladapt Immunotherapeutics, Inc. Compositions and methods for identification of cross-reactive allergenic proteins and treatment of allergies
US20240041978A1 (en) 2021-03-03 2024-02-08 The U.S.A., As Represented By The Secretary, Department Of Health And Human Services La protein as a novel regulator of osteoclastogenesis
JP2024511373A (en) 2021-03-18 2024-03-13 ノバルティス アーゲー Biomarkers and their use for cancer
WO2022204581A2 (en) 2021-03-26 2022-09-29 Scholar Rock, Inc. Tgf-beta inhibitors and use thereof
TW202304979A (en) 2021-04-07 2023-02-01 瑞士商諾華公司 USES OF ANTI-TGFβ ANTIBODIES AND OTHER THERAPEUTIC AGENTS FOR THE TREATMENT OF PROLIFERATIVE DISEASES
KR20240004462A (en) 2021-04-08 2024-01-11 마렝고 테라퓨틱스, 인크. Multifunctional molecules that bind to TCR and their uses
WO2022235929A1 (en) 2021-05-05 2022-11-10 Radius Pharmaceuticals, Inc. Animal model having homologous recombination of mouse pth1 receptor
CA3216320A1 (en) 2021-05-18 2022-11-24 Abbott Laboratories Methods of evaluating brain injury in a pediatric subject
WO2022256723A2 (en) 2021-06-03 2022-12-08 Scholar Rock, Inc. Tgf-beta inhibitors and therapeutic use thereof
WO2022261018A1 (en) 2021-06-07 2022-12-15 Providence Health & Services - Oregon Cxcr5, pd-1, and icos expressing tumor reactive cd4 t cells and their use
WO2022261183A2 (en) 2021-06-08 2022-12-15 Dana-Farber Cancer Institute, Inc. Compositions and methods for treating and/or identifying an agent for treating intestinal cancers
CA3222291A1 (en) 2021-06-14 2022-12-22 Jaime MARINO Methods of diagnosing or aiding in diagnosis of brain injury caused by acoustic energy, electromagnetic energy, an over pressurization wave, and/or blast wind
CN113403690B (en) * 2021-06-21 2022-07-19 吉林大学 DNA coding compound library drug molecule fishing method
AU2022299185A1 (en) 2021-06-23 2024-01-25 Scholar Rock, Inc. A myostatin pathway inhibitor in combination with a glp-1 pathway activator for use in treating metabolic disorders
IL309957A (en) 2021-07-14 2024-03-01 2Seventy Bio Inc Engineered t cell receptors fused to binding domains from antibodies
IL310535A (en) 2021-08-10 2024-03-01 Byomass Inc Anti-gdf15 antibodies, compositions and uses thereof
WO2023025927A1 (en) 2021-08-26 2023-03-02 Glycanostics S.R.O Glycoprotein biomarkers for diagnosing cancer
AU2022339759A1 (en) 2021-08-31 2024-03-07 Abbott Laboratories Methods and systems of diagnosing brain injury
WO2023041565A1 (en) 2021-09-14 2023-03-23 Glycanostics S.R.O Use of lectins to determine mammaglobin-a glycoforms in breast cancer
WO2023044483A2 (en) 2021-09-20 2023-03-23 Voyager Therapeutics, Inc. Compositions and methods for the treatment of her2 positive cancer
WO2023044094A1 (en) 2021-09-20 2023-03-23 Alnylam Pharmaceuticals, Inc. Inhibin subunit beta e (inhbe) modulator compositions and methods of use thereof
WO2023056268A1 (en) 2021-09-30 2023-04-06 Abbott Laboratories Methods and systems of diagnosing brain injury
WO2023069421A1 (en) 2021-10-18 2023-04-27 Byomass Inc. Anti-activin a antibodies, compositions and uses thereof
WO2023092004A1 (en) 2021-11-17 2023-05-25 Voyager Therapeutics, Inc. Compositions and methods for the treatment of tau-related disorders
WO2023097254A1 (en) 2021-11-24 2023-06-01 Visterra, Inc. Engineered antibody molecules to cd138 and uses thereof
WO2023097119A2 (en) 2021-11-29 2023-06-01 Dana-Farber Cancer Institute, Inc. Methods and compositions to modulate riok2
TW202333772A (en) 2021-12-01 2023-09-01 美商威特拉公司 Methods of using interleukin-2 agents
WO2023114978A1 (en) 2021-12-17 2023-06-22 Abbott Laboratories Systems and methods for determining uch-l1, gfap, and other biomarkers in blood samples
WO2023122213A1 (en) 2021-12-22 2023-06-29 Byomass Inc. Targeting gdf15-gfral pathway cross-reference to related applications
WO2023118508A1 (en) 2021-12-23 2023-06-29 Bavarian Nordic A/S Recombinant mva viruses for intraperitoneal administration for treating cancer
US20230213536A1 (en) 2021-12-28 2023-07-06 Abbott Laboratories Use of biomarkers to determine sub-acute traumatic brain injury (tbi) in a subject having received a head computerized tomography (ct) scan that is negative for a tbi or no head ct scan
WO2023147107A1 (en) 2022-01-31 2023-08-03 Byomass Inc. Myeloproliferative conditions
WO2023150652A1 (en) 2022-02-04 2023-08-10 Abbott Laboratories Lateral flow methods, assays, and devices for detecting the presence or measuring the amount of ubiquitin carboxy-terminal hydrolase l1 and/or glial fibrillary acidic protein in a sample
TW202342548A (en) 2022-02-07 2023-11-01 美商威特拉公司 Anti-idiotype antibody molecules and uses thereof
TW202400637A (en) 2022-04-25 2024-01-01 美商威特拉公司 Antibody molecules to april and uses thereof
WO2023220695A2 (en) 2022-05-13 2023-11-16 Voyager Therapeutics, Inc. Compositions and methods for the treatment of her2 positive cancer
EP4296279A1 (en) 2022-06-23 2023-12-27 Fraunhofer-Gesellschaft zur Förderung der angewandten Forschung e.V. Anti-transthyretin (ttr) binding proteins and uses thereof
WO2024006876A1 (en) 2022-06-29 2024-01-04 Abbott Laboratories Magnetic point-of-care systems and assays for determining gfap in biological samples
WO2024030976A2 (en) 2022-08-03 2024-02-08 Voyager Therapeutics, Inc. Compositions and methods for crossing the blood brain barrier
WO2024059708A1 (en) 2022-09-15 2024-03-21 Abbott Laboratories Biomarkers and methods for differentiating between mild and supermild traumatic brain injury
WO2024062038A1 (en) 2022-09-21 2024-03-28 Elthera Ag Novel binding molecules binding to l1cam

Family Cites Families (71)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPS53133684A (en) * 1977-04-26 1978-11-21 Noda Sangyo Kagaku Kenkyusho Novel bacteriophage and fabricating same
DE2814039C3 (en) * 1978-03-31 1981-02-19 Gesellschaft Fuer Biotechnologische Forschung Mbh (Gbf), 3300 Braunschweig Process for the production of hybrid bacteria
US4411994A (en) * 1978-06-08 1983-10-25 The President And Fellows Of Harvard College Protein synthesis
JPS5558096A (en) * 1978-10-25 1980-04-30 Noda Sangyo Kagaku Kenkyusho Method of making novel recombined dna
JPS5561798A (en) * 1978-10-30 1980-05-09 Noda Sangyo Kagaku Kenkyusho Preparation of novel recombination dna
US4528266A (en) * 1979-10-01 1985-07-09 George Pieczenik Method of inserting unique DNA sequences into DNA vectors
US4359535A (en) * 1979-10-01 1982-11-16 George Pieczenik Autonomously replicating DNA containing inserted DNA sequences
US4769326A (en) 1980-02-29 1988-09-06 The Regents Of The University Of California Expression linkers
US4338397A (en) * 1980-04-11 1982-07-06 President And Fellows Of Harvard College Mature protein synthesis
DE3126021A1 (en) * 1981-07-02 1983-07-28 Basf Ag, 6700 Ludwigshafen METHOD FOR PRODUCING OPTICALLY PURE D- OR L-LACTIC ACID
US4603112A (en) * 1981-12-24 1986-07-29 Health Research, Incorporated Modified vaccinia virus
US4593002A (en) 1982-01-11 1986-06-03 Salk Institute Biotechnology/Industrial Associates, Inc. Viruses with recombinant surface proteins
EP0088994B1 (en) * 1982-03-15 1991-06-19 Schering Corporation Hybrid dna, binding composition prepared thereby and processes therefor
US4508826A (en) * 1982-04-15 1985-04-02 Merck & Co., Inc. Bacteriophage DNA cloning vector TG1 and microorganisms containing TG1
US4595658A (en) * 1982-09-13 1986-06-17 The Rockefeller University Method for facilitating externalization of proteins synthesized in bacteria
US4757013A (en) * 1983-07-25 1988-07-12 The Research Foundation Of State University Of New York Cloning vehicles for polypeptide expression in microbial hosts
AU560584B2 (en) * 1983-07-28 1987-04-09 Bayer Aktiengesellschaft Homologues of aprotinin
DE3485850T2 (en) * 1983-11-08 1993-01-21 Teijin Ltd GENE FRAGMENTS DERIVED FROM THE HUMAN IMMUNOGLOBULIN GENE.
US4703004A (en) * 1984-01-24 1987-10-27 Immunex Corporation Synthesis of protein with an identification peptide
US4797363A (en) * 1984-03-19 1989-01-10 Board Of Trustees, University Of Illinois Bacteriophages as recognition and identification agents
JPS61104788A (en) * 1984-10-26 1986-05-23 Teijin Ltd Nucleic acid base sequence
JPS61134325A (en) 1984-12-04 1986-06-21 Teijin Ltd Expression of hybrid antibody gene
US4774180A (en) * 1986-02-26 1988-09-27 Massachusetts Institute Of Technology Construction and application of polyproteins
US4683202A (en) 1985-03-28 1987-07-28 Cetus Corporation Process for amplifying nucleic acid sequences
AU5668486A (en) * 1985-03-29 1986-10-23 Biotechnica International Inc. Secretion vector
DE3546807C2 (en) 1985-03-30 1991-03-28 Marc Genf/Geneve Ch Ballivet
US5866363A (en) * 1985-08-28 1999-02-02 Pieczenik; George Method and means for sorting and identifying biological information
IL79881A (en) 1985-08-28 1995-12-08 Pieczenik George Populations of random oligonucleotide coding sequences and peptides
US4908773A (en) * 1987-04-06 1990-03-13 Genex Corporation Computer designed stabilized proteins and method for producing same
GB2188322A (en) 1986-03-26 1987-09-30 Bayer Ag Aprotinin and analogues thereof produced by a recombinant host
US5225539A (en) * 1986-03-27 1993-07-06 Medical Research Council Recombinant altered antibodies and methods of making altered antibodies
GB2188933A (en) 1986-04-10 1987-10-14 Bayer Ag Expression vectors for production of polypeptides, method for enhanced expression of polypeptides, hosts containing the expression vectors, products manufactured thereby
US4829052A (en) * 1986-06-11 1989-05-09 Monsanto Company Serine protease inhibitors
ATE87659T1 (en) 1986-09-02 1993-04-15 Enzon Lab Inc BINDING MOLECULES WITH SINGLE POLYPEPTIDE CHAIN.
US4704692A (en) * 1986-09-02 1987-11-03 Ladner Robert C Computer based system and method for determining and displaying possible chemical structures for converting double- or multiple-chain polypeptides to single-chain polypeptides
IL85035A0 (en) 1987-01-08 1988-06-30 Int Genetic Eng Polynucleotide molecule,a chimeric antibody with specificity for human b cell surface antigen,a process for the preparation and methods utilizing the same
WO1988006601A1 (en) 1987-03-02 1988-09-07 Genex Corporation Gene repressors
DE3852304T3 (en) 1987-03-02 1999-07-01 Enzon Lab Inc Organism as carrier for "Single Chain Antibody Domain (SCAD)".
ATE135403T1 (en) 1987-03-10 1996-03-15 New England Biolabs Inc PREPARATION AND PURIFICATION OF A PROTEIN FUSIONED WITH A BINDING PROTEIN
EP0285123A3 (en) 1987-04-03 1989-02-01 Stabra AG A method for complete mutagenesis of nucleic acids
DE3724570A1 (en) 1987-06-27 1989-01-05 Bayer Ag HUMAN APROTININ, WHOSE LYS REST IN POSITION 15 IS REPLACED BY ANOTHER PROTOGENIC AMINO ACID REST
GB2208511A (en) 1987-08-07 1989-04-05 Bayer Ag Variants of bovine pancreatic trypsin inhibitor produced by recombinant dna technology
DK450187D0 (en) 1987-08-28 1987-08-28 Novo Industri As PROCEDURE FOR THE PRODUCTION OF PROTEINS
DK225488D0 (en) 1988-04-26 1988-04-26 Novo Industri As POLYPEPTIDE
DE3731874A1 (en) * 1987-09-18 1989-03-30 Schering Ag METHOD FOR THE PRODUCTION OF PEPTIDES BY SPECIFIC CUTTING OF GENETICALLY OBTAINED FUSION PROTEINS WITH COLLAGENASES
DK172110B1 (en) 1988-04-15 1997-10-27 Gen Hospital Corp Method for Isolating Mutants of DNA Sequences and Their Use in Identifying Cell Surface Proteins
US5010175A (en) * 1988-05-02 1991-04-23 The Regents Of The University Of California General method for producing and selecting peptides with specific properties
DE68927933T2 (en) * 1988-09-02 1997-08-14 Dyax Corp PRODUCTION AND SELECTION OF RECOMBINANT PROTEINS WITH DIFFERENT BINDING POINTS
US5223409A (en) * 1988-09-02 1993-06-29 Protein Engineering Corp. Directed evolution of novel binding proteins
US6291158B1 (en) * 1989-05-16 2001-09-18 Scripps Research Institute Method for tapping the immunological repertoire
US7413537B2 (en) * 1989-09-01 2008-08-19 Dyax Corp. Directed evolution of disulfide-bonded micro-proteins
US5747334A (en) * 1990-02-15 1998-05-05 The University Of North Carolina At Chapel Hill Random peptide library
US5498538A (en) * 1990-02-15 1996-03-12 The University Of North Carolina At Chapel Hill Totally synthetic affinity reagents
US5427908A (en) 1990-05-01 1995-06-27 Affymax Technologies N.V. Recombinant library screening methods
EP0600866B1 (en) 1990-06-01 1997-12-03 Chiron Corporation Compositions and methods for identifying biologically active molecules
US5723286A (en) * 1990-06-20 1998-03-03 Affymax Technologies N.V. Peptide library and screening systems
GB9015198D0 (en) * 1990-07-10 1990-08-29 Brien Caroline J O Binding substance
AU8740491A (en) * 1990-09-28 1992-04-28 Protein Engineering Corporation Proteinaceous anti-dental plaque agents
IL99553A0 (en) 1990-09-28 1992-08-18 Ixsys Inc Compositions containing oligonucleotides linked to expression elements,a kit for the preparation of vectors useful for the expression of a diverse population of random peptides and methods utilizing the same
IL99552A0 (en) 1990-09-28 1992-08-18 Ixsys Inc Compositions containing procaryotic cells,a kit for the preparation of vectors useful for the coexpression of two or more dna sequences and methods for the use thereof
JP3298879B2 (en) 1990-12-20 2002-07-08 イグジス,インコーポレイテッド Optimization of binding proteins
ATE431364T1 (en) 1991-03-01 2009-05-15 Dyax Corp INHIBITORS OF HUMAN NEUTROPHIL ELASTASIS
DK1279731T3 (en) 1991-03-01 2007-09-24 Dyax Corp Process for the development of binding mini-proteins
KR20030096423A (en) * 1992-03-06 2003-12-31 엔. 브이. 이노제네틱스 소시에떼아노님 Process for the determination of peptides corresponding to immunologically important epitopes and their use in a process for determination of antibodies or biotinylated peptides corresponding to immunologically important epitopes, a process for preparing them and compositions containing them
US5858679A (en) * 1992-11-12 1999-01-12 Fornace, Jr.; Albert J. Method for determining the presence of functional p53 by measuring GADD45 protein expression
US5679548A (en) 1993-02-02 1997-10-21 The Scripps Research Institute Methods for producing polypeptide metal binding sites and compositions thereof
US5432155A (en) * 1993-06-29 1995-07-11 The Salk Institute For Biological Studies Conotoxins I
ATE310812T1 (en) 1994-01-18 2005-12-15 Scripps Research Inst DERIVATIVES OF ZINC FINGER PROTEINS AND METHODS
JP4118327B2 (en) 1994-08-20 2008-07-16 ゲンダック・リミテッド Improvements in or related to binding proteins for DNA recognition
US5789538A (en) * 1995-02-03 1998-08-04 Massachusetts Institute Of Technology Zinc finger proteins with high affinity new DNA binding specificities
US5955582A (en) * 1997-09-26 1999-09-21 Beckman Coulter, Inc. Antibody against a 3-aminophenylboronic-glycated protein complex and its use in an immunoassay

Also Published As

Publication number Publication date
ATE478949T1 (en) 2010-09-15
DK1820858T3 (en) 2009-11-02
US7893007B2 (en) 2011-02-22
ES2219638T3 (en) 2004-12-01
ATE262036T1 (en) 2004-04-15
DK1279731T3 (en) 2007-09-24
EP0575485A1 (en) 1993-12-29
DE69233769D1 (en) 2009-09-24
EP0573611B9 (en) 2004-10-06
DE69233325T2 (en) 2005-03-10
JP4146512B2 (en) 2008-09-10
ATE439435T1 (en) 2009-08-15
DK1452599T3 (en) 2010-12-13
ES2287206T3 (en) 2007-12-16
AU1545692A (en) 1992-10-06
EP1452599B1 (en) 2010-08-25
JP3447731B2 (en) 2003-09-16
ATE363532T1 (en) 2007-06-15
EP2224001A1 (en) 2010-09-01
EP0573611B1 (en) 2004-03-17
CA2105300C (en) 2008-12-23
DK0573611T3 (en) 2004-07-19
JP4451611B2 (en) 2010-04-14
DE69233697D1 (en) 2007-07-12
JPH07501923A (en) 1995-03-02
WO1992015677A1 (en) 1992-09-17
AU1578792A (en) 1992-10-06
JP4618570B2 (en) 2011-01-26
CA2105300A1 (en) 1992-09-02
DE69233325D1 (en) 2004-04-22
ES2330052T3 (en) 2009-12-03
EP1452599A1 (en) 2004-09-01
EP1279731A1 (en) 2003-01-29
EP1820858A1 (en) 2007-08-22
EP1279731B1 (en) 2007-05-30
JP2004000221A (en) 2004-01-08
JP2008260770A (en) 2008-10-30
DE69233697T2 (en) 2008-01-24
WO1992015679A1 (en) 1992-09-17
US20090234101A1 (en) 2009-09-17
JP2010183908A (en) 2010-08-26
DE69233795D1 (en) 2010-10-07
ES2351693T3 (en) 2011-02-09
JP2003159086A (en) 2003-06-03
EP0573611A1 (en) 1993-12-15
EP1820858B1 (en) 2009-08-12
JPH07501203A (en) 1995-02-09

Similar Documents

Publication Publication Date Title
CA2105303A1 (en) Display phage
US5837500A (en) Directed evolution of novel binding proteins
US20070259417A1 (en) Directed evolution of novel binding proteins
US7413537B2 (en) Directed evolution of disulfide-bonded micro-proteins
CA1340288C (en) Generation and selection of novel binding proteins
IL120939A (en) Method for obtaining nucleic acid encoding a proteinaceous binding domain

Legal Events

Date Code Title Description
EEER Examination request
FZDE Discontinued