AU2006203197B2 - Derivatives of growth hormone and related proteins - Google Patents

Derivatives of growth hormone and related proteins Download PDF

Info

Publication number
AU2006203197B2
AU2006203197B2 AU2006203197A AU2006203197A AU2006203197B2 AU 2006203197 B2 AU2006203197 B2 AU 2006203197B2 AU 2006203197 A AU2006203197 A AU 2006203197A AU 2006203197 A AU2006203197 A AU 2006203197A AU 2006203197 B2 AU2006203197 B2 AU 2006203197B2
Authority
AU
Australia
Prior art keywords
leu
ser
ala
thr
arg
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.)
Ceased
Application number
AU2006203197A
Other versions
AU2006203197A1 (en
Inventor
George N Cox III
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.)
Bolder Biotechnology Inc
Original Assignee
Bolder Biotechnology Inc
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
Priority claimed from AU2002306305A external-priority patent/AU2002306305C1/en
Application filed by Bolder Biotechnology Inc filed Critical Bolder Biotechnology Inc
Priority to AU2006203197A priority Critical patent/AU2006203197B2/en
Publication of AU2006203197A1 publication Critical patent/AU2006203197A1/en
Application granted granted Critical
Publication of AU2006203197B2 publication Critical patent/AU2006203197B2/en
Priority to AU2008203051A priority patent/AU2008203051B2/en
Anticipated expiration legal-status Critical
Ceased legal-status Critical Current

Links

Description

P001 Section 29 Regulation 3.2(2)
AUSTRALIA
Patents Act 1990 COMPLETE SPECIFICATION STANDARD PATENT Application Number: Lodged: Invention Title: Derivatives of growth hormone and related proteins The following statement is a full description of this invention, including the best method of performing it known to us: DERIVATIVES OF GROWTH HORMONE AND RELATED PROTEINS Field of the Invention The present invention relates to genetically engineered therapeutic proteins. More specifically, the engineered proteins include growth hormone and related proteins.
Background of the Invention The following proteins are encoded by genes of the growth hormone (GH) supergene family (Bazan (1990); Mott and Campbell (1995); Silvennoinen and Ihle (1996)): growth hormone, prolactin, placental lactogen, erythropoietin (EPO), thrombopoietin (TPO), interleukin-2 IL-3, IL-4, IL-5, IL-6, IL-7, IL-9, IL-10, IL-11, IL-12 (p35 subunit), IL- 13, IL-15, oncostatin M, ciliary neurotrophic factor, leukemia inhibitory factor, alpha interferon, beta interferon, gamma interferon, omega interferon, tau interferon, granulocytecolony stimulating factor (G-CSF), granulocyte-macrophage colony stimulating factor (GM- CSF), macrophage colony stimulating factor (M-CSF) and cardiotrophin-1 (CT-I) ("the GH supergene family"). It is anticipated that additional members of this gene family will be identified in the future through gene cloning and sequencing. Members of the GH supergene family have similar secondary and tertiary structures, despite the fact that they generally have limited amino acid or DNA sequence identity. The shared structural features allow new members of the gene family to be readily identified.
There is considerable interest on the part of patients and healthcare providers in the development of long acting, "user-friendly" protein therapeutics. Proteins are expensive to manufacture and, unlike conventional small molecule drugs, are not readily absorbed by the body. Moreover, they are digested if taken orally. Therefore, natural proteins must be administered by injection. After injection, most proteins are cleared rapidly from the body, necessitating frequent, often daily, injections. Patients dislike injections, which leads to reduced compliance and reduced drug efficacy. Some proteins, such as erythropoietin (EPO), are effective when administered less often (three times per week for EPO) because they are glycosylated. However, glycosylated proteins are produced using expensive mammalian cell expression systems.
The length of time an injected protein remains in the body is finite and is determined by, the protein's size and whether or not the protein contains covalent modifications such
I
as glycosylation. Circulating concentrations of injected proteins change constantly, often by C several orders of magnitude, over a 24-hour period. Rapidly changing concentrations of protein agonists can have dramatic downstream consequences, at times under-stimulating and at other times over-stimulating target cells. Similar problems plague protein antagonists.
These fluctuations can lead to decreased efficacy and increased frequency of adverse side 0 effects for protein therapeutics. The rapid clearance of recombinant proteins from the body D0 significantly increases the amount of protein required per patient and dramatically increases Sthe cost of treatment The cost of human protein pharmaceuticals is expected to increase dramatically in the years ahead as new and existing drugs are approved for more disease indications.
Thus, there is a need to develop protein delivery technologies that lower the costs of protein therapeutics to patients and healthcare providers. The present invention provides a solution to this problem by providing methods to prolong the circulating half-lives of protein therapeutics in the body so that the proteins do not have to be injected frequently. This solution also satisfies the needs and desires of patients for protein therapeutics that are userfriendly", protein therapeutics that do not require frequent injections. The present invention solves these and other problems by providing biologically active, cysteine-added variants of members of the growth hormone supergene family. The invention also provides for the chemical modification of these variants with cysteine-reactive polymers or other types of cysteine-reactive moieties to produce derivatives thereof and the molecules so produced.
Summary of the Invention The present invention provides cysteine variants of members of the GH supergene family. The variants comprise a cysteine residue substituted for a nonessential amino acid of the proteins. Preferably, the variants comprise a cysteine residue substituted for an amino acid selected from amino acids in the loop regions, the ends of the alpha helices, proximal to the first amphipathic helix, and distal to the final amphipathic helix or wherein the cysteine residue is added at the N-terminus or C-terminus of the proteins. Preferred sites for substitution are the N- and O-linked glycosylation sites.
Also provided are cysteine variants wherein the amino acid substituted for is in the A- B loop, B-C loop, the C-D loop or D-E loop of interferon/interferon-I 0-like members of the GH supergene family.
I Also provided are cysteine variants of members of the GH supergene family wherein the cysteine residue is introduced between two amino acids in the natural protein. In particular, the cysteine residue is introduced into the loop regions, the ends of the alpha Shelices, proximal to the first amphipathic helix, or distal to the final amphipathic helix. Even M 5 more particularly, the cysteine variant is introduced between two amino acids in an N-O- *C linked glycosylation site or adjacent to an amino acid in an N-linked or O-linked 0 glycosylation site.
C More particularly are provided cysteine variants wherein the loop region where the cysteine is introduced is the A-B loop, the B-C loop, the C-D loop or D-E loop of interferon/interferon-10-like members of the GH supergene family.
Such cysteine substitutions or insertion mutations also can include the insertion of one or more additional amino acids amino acids at the amino-terminal or carboxy-terminal to the cysteine substitution or insertion.
Also provided are cysteine variants that are further derivatised by PEGylating the cysteine variants and including the derivatised proteins produced thereby.
As set forth in the examples, specific cysteine variants of the members of the GH supergene family also are provided, including for example, variants of GH. The GH cysteine variants can have the substituted-for amino acid or inserted cysteine located at the N-terminal end of the A-B loop, the B-C loop, the C-D loop, the first three or last three amino acids in the A, B, C and D helices and the amino acids proximal to helix A and distal to helix D.
More particularly, the cysteine can be substituted for the following amino acids: FI, T3, PS, E33, A34, K38, E39, K40, S43, Q46, N47, P48, Q49, T50, S51, S55, T60, A98, N99, S100, G104, Al05, S106, E129, D130, 0131, S132, P133, T135, G136, Q137, K140, Q141, T142, S144, K145, D147, T148, N149, S150, H151, N152, D153, S184, E186, G187, S188, and 0190.
Other examples of cysteine variants according to the invention include erythropoietin variants. Erythropoietin variants include those wherein the substituted for amino acid is located in the A-B loop, the B-C loop, the C-D loop, the amino acids proximal to helix A and distal to helix D and the N- or C-terminus. Even more specifically, the EPO cysteine variants include molecules wherein the amino acids indicated below have a cysteine substituted therefor: serine-126, N24, 125, T26, N38, 139, T40, N83, S84, Al, P2, P3, R4, D8, S9, T27, G28, A30, E31, H32, S34, N36, D43, T44, K45, N47, A50, K52, E55, G57, Q58, G77, Q78, D A79, Q86, W88, E89, T107, R110, AllI, G113, A114, Q115, K116, E117, A118, S120, P121, P122, D123, A124, A125, A127, A128, T132, K154, T157, G158, E159, A160, T163, G164, D165, R166 and The members of the GH supergene family include growth hormone, prolactin, c 5 placental lactogen, erythropoietin, thrombopoietin, interleukin-2, interleukin-3, interleukin-4, interleukin-6, interleukin-7, interleukin-9, interleukin-10, interleukin-1 1, 0 interleukin-12 (p35 subunit), interleukin-13, interleukin-15, oncostatin M, ciliary C1 neurotrophic factor, leukemia inhibitory factor, alpha interferon, beta interferon, gamma interferon, omega interferon, tau interferon, granulocyte-colony stimulating factor, lo granulocyte-macrophage colony stimulating factor, macrophage colony stimulating factor, cardiotrophin-1 and other proteins identified and classified as members of the family. The proteins can be derived from any animal species including human, companion animals and farm animals.
Other variations and modifications to the invention will be obvious to those skilled in the art based on the specification and the "rules" set forth herein. All of these are considered as part of the invention.
Detailed Description of the Invention The present invention relates to cysteine variants and, among other things, the sitespecific conjugation of such proteins with polyethylene glycol (PEG) or other such moieties.
PEG is a non-antigenic, inert polymer that significantly prolongs the length of time a protein circulates in the body. This allows the protein to be effective for a longer period of time.
Covalent modification of proteins with PEG has proven to be a useful method to extend the circulating half-lives of proteins in the body (Abuchowski et al., 1984; Hershfield, 1987; Meyers et al., 1991). Covalent attachment of PEG to a protein increases the protein's effective size and reduces its rate of clearance rate from the body. PEGs are commercially available in several sizes, allowing the circulating half-lives ofPEG-modified proteins to be tailored for individual indications through use of different size PEGs. Other benefits of PEG modification include an increase in protein solubility, an increase in in vivo protein stability and a decrease in protein immunogenicity (Katre et al., 1987; Katre, 1990).
The preferred method for PEGylating proteins is to covalently attach PEG to cysteine residues using cysteine-reactive PEGs. A number of highly specific, cysteine-reactive PEGs ND with different reactive groups maleimide, vinylsulfone) and different size PEGs (2-20 kDa) are commercially available from Shearwater, Polymers, Inc., Huntsville, AL). At neutral pH, these PEG reagents selectively attach to "free" cysteine residues, cysteine residues not involved in disulfide bonds. The conjugates are hydrolytically stable. Use of cysteine-reactive PEGs allows the development of homogeneous PEG-protein conjugates of C1 defined structure.
O Considerable progress has been made in recent years in determining the structures of C' commercially important protein therapeutics and understanding how they interact with their protein targets, cell-surface receptors, proteases, etc. This structural information can be o0 used to design PEG-protein conjugates using cysteine-reactive PEGs. Cysteine residues in most proteins participate in disulfide bonds and are not available for PEGylation using cysteine-reactive PEGs. Through in vitro mutagenesis using recombinant DNA techniques, additional cysteine residues can be introduced anywhere into the protein. The added cysteines can be introduced at the beginning of the protein, at the end of the protein, between two amino acids in the protein sequence or, preferably, substituted for an existing amino acid in the protein sequence. The newly added "free" cysteines can serve as sites for the specific attachment of a PEG molecule using cysteine-reactive PEGs. The added cysteine must be exposed on the protein's surface and-accessible for PEGylation for this method to be successful. If the site used to introduce an added cysteine site is non-essential for biological activity, then the PEGylated protein will display essentially wild type (normal) in vitro bioactivity. The major technical challenge in PEGylating proteins with cysteine-reactive PEGs is the identification of surface exposed, non-essential regions in the target protein where cysteine residues can be added or substituted for existing amino acids without loss of bioactivity.
Cysteine-added variants of a few human proteins and PEG-polymer conjugates of these proteins have been described. US patent 5,206,344 describes cysteine-added variants of IL-2. These cysteine-added variants are located within the first 20 amino acids from the amino terminus of the mature IL-2 polypeptide chain. The preferred cysteine variant is at position 3 of the mature polypeptide chain, which corresponds to a threonine residue that is O-glycosylated in the naturally occurring protein. Substitution of cysteine for threonine at position 3 yields an IL-2 variant that can be PEGylated with a cysteine-reactive PEG and retain full in vitro bioactivity (Goodson and Katre, 1990). In contrast, natural IL-2 PEGylated with lysine-reactive PEGs displays reduced in itro bioactivity (Goodson and Katre, 1990).
F- The effects of cysteine substitutions at other positions in IL-2 were not reported.
SUS patent 5,166,322 teaches cysteine-added variants of IL-3. These variants are located within the first 14 amino acids from the N-terminus of the mature protein sequence.
The patent teaches expression of the proteins in bacteria and covalent modification of the proteins with cysteine-reactive PEGs. No information is provided as to whether the cysteineadded variants and PEG-conjugates of IL-3 are biologically active. Cysteine-added variants Sat other positions in the polypeptide chain were not reported.
O World patent application W09412219 and PCT application US95/06540 teach C 10 cysteine-added variants of insulin-like growth factor-I (IGF-I). IGF-I has a very different structure from GH and is not a member of the GH supergene family (Mott and Campbell, 1995). Cysteine substitutions at many positions in the IGF-I protein are described. Only certain of the cysteine-added variants are biologically active. The preferred site for the cysteine added variant is at amino acid position 69 in the mature protein chain. Cysteine substitutions at positions near the N-terminus of the protein (residues 1-3) yielded IGF-I variants with reduced biological activities and improper disulfide bonds.
World patent application W09422466 teaches two cysteine-added variants of insulinlike growth factor (IGF) binding protein-1, which has a very different structure than GH and is not a member of the GH supergene family. The two cysteine-added IGF binding protein-1 variants disclosed are located at positions 98 and 101 in the mature protein chain and correspond to serine residues that are phosphorylated in the naturally-occurring protein.
US patent application 07/822296 teaches cysteine added variants of tumor necrosis factor binding protein, which is a soluble, truncated form of the tumor necrosis factor cellular receptor. Tumor necrosis factor binding protein has a very different structure than GH and is not a member of the GH supergene family.
IGF-I, IGF binding protein-1 and tumor necrosis factor binding protein have secondary and tertiary structures that are very different from GH and the proteins are not members of the GH supergene family. Because of this, it is difficult to use the information gained from studies of IGF-I, IGF binding protein-1 and tumor necrosis factor binding protein to create cysteine-added variants of members of the GH supergene family. The studies with IL-2 and IL-3 were carried out before the structures of IL-2 and IL-3 were known (McKay 1992; Bazan, 1992) and before it was known that these proteins are members of the GH supergene family. Previous experiments aimed at identifying preferred sites for adding -s cysteine residues to IL-2 and IL-3 were largely empirical and were performed prior to experiments indicating that members of the GH supergene family possessed similar secondary and tertiary structures.
5 Based on the structural information now available for members of the GH supergene Sfamily, the present invention provides "rules" for determining ariori which regions and amino acid residues in members of the GH supergene family can be used to introduce or N substitute cysteine residues without significant loss of biological activity. In contrast to the naturally occurring proteins, these cysteine-added variants of members of the GH supergene C o1 family will possess novel properties such as the ability to be covalently modified at defined sites within the polypeptide chain with cysteine-reactive polymers or other types of cysteinereactive moieties. The covalently modified proteins will be biologically active.
GH is the best-studied member of the GH supergene family. GH is a 22 kDa protein secreted by the pituitary gland. GH stimulates metabolism of bone, cartilage and muscle and s1 is the body's primary hormone for stimulating somatic growth during childhood.
Recombinant human GH (rhGH) is used to treat short stature resulting from GH inadequacy and renal failure in children. GH is not glycosylated and can be produced in a fully active form in bacteria. The protein has a short in vivo half-life and must be administered by daily subcutaneous injection for maximum effectiveness (MacGillivray et al., 1996). Recombinant human GH (rhGH) was approved recently for treating cachexia in AIDS patients and is under study for treating cachexia associated with other diseases.
The sequence of human GH is well known (see, Martial et al. 1979; Goeddel et al. 1979 which are incorporated herein by reference; SEQ ID NO: GH is closely related in sequence to prolactin and placental lactogen and these three proteins were considered originally to comprise a small gene family. The primary sequence of GH is highly conserved among animal species (Abdel-Meguid et al., 1987), consistent with the protein's broad species cross-reactivity. The three dimensional folding pattern of porcine GH has been solved by X-ray crystallography (Abdel-Meguid et al., 1987). The protein has a compact globular structure, comprising four amphipathic alpha helical bundles joined by loops.
Human GH has a similar structure (de Vos et al., 1992). The four alpha helical regions are termed A-D beginning from the N-terminus of the protein. The loop regions are referred to by the helical regions they join, the A-B loop joins helical bundles A and B. The A-B and C-D loops are long, whereas the B-C loop is short. GH contains four cysteine residues, all of which participate in disulfide bonds. The disulfide assignments are cysteine53 joined to ND cysteinel65 and cysteinel 82 joined to cysteinel89.
The crystal structure of GH bound to its receptor revealed that GH has two receptor s binding sites and binds two receptor molecules (Cunningham et al., 1991; de Vos et al., 1992). The two receptor binding sites are referred to as site I and site II. Site 1 encompasses the Carboxy (C)-terminal end of helix D and parts of helix A and the A-B loop, whereas site SII encompasses the Amino (N)-terminal region of helix A and a portion of helix C. Binding Sof GH to its receptor occurs sequentially, with site I always binding first. Site 11 then engages C" 10 a second GH receptor, resulting in receptor dimerization and activation of the intracellular signaling pathways that lead to cellular responses to GH. A GH mutein in which site 1 has been mutated (a glycine to arginine mutation at amino acid 120) is able to bind a single GH receptor, but is unable to dimerize GH receptors; this mutein acts as a GH antagonist in vitro, presumably by occupying GH receptor sites without activating intracellular signaling pathways (Fuh et al., 1992).
The roles of particular regions and amino acids in GH receptor binding and intracellular signaling also have been studied using techniques such as mutagenesis, monoclonal antibodies and proteolytic digestion. The first mutagenesis experiments entailed replacing entire domains of GH with similar regions of the closely related protein, prolactin (Cunningham et al., 1989). One finding was that replacement of the B-C loop of GH with that of prolactin did not affect binding of the hybrid GH protein to a soluble form of the human GH receptor, implying that the B-C loop was non-essential for receptor binding.
Alanine scanning mutagenesis (replacement of individual amino acids with alanine) identified 14 amino acids that are critical for GH bioactivity (Cunningham and Wells, 1989). These amino acids are located in the helices A, B, C, and D and the A-B loop and correspond to sites I and II identified from the structural studies. Two lysine residues at amino acid positions 41 and 172, K41 and K172, were determined to be critical components of the site I receptor binding site, which explains the decrease in bioactivity observed when K172 is acetylated (Teh and Chapman, 1988). Modification of K168 also significantly reduced GH receptor binding and bioactivity (de la Llosa et al., 1985; Martal et al., 1985; Teh and Chapman, 1988). Regions of GH responsible for binding the GH receptor have also been studied using monoclonal antibodies (Cunningham et al., 1989). A series of eight monoclonal antibodies was generated to human GH and analyzed for the ability to neutralize GH activity and prevent binding of GH to its recombinant soluble receptor. The latter studies N allowed the putative binding site for each monoclonal antibody to be localized within the GH three-dimensional structure. Of interest was that monoclonal antibodies 1 and 8 were unable to displace GH from binding its receptor. The binding sites for these monoclonal antibodies were localized to the B-C loop (monoclonal number 1) and the N-terminal end of the A-B 0 loop (monoclonal number No monoclonals were studied that bound the C-D loop n specifically. The monoclonal antibody studies suggest that the B-C loop and N-terminal end Sof the A-B loop are non-essential for receptor binding. Finally, limited cleavage of GH with trypsin was found to produce a two chain derivative that retained full activity (Mills et al., 1980; Li, 1982). Mapping studies indicated that trypsin cleaved and/or deleted amino acids between positions 134 and 149, which corresponds to the C-D loop. These studies suggest the C-D loop is not involved in receptor binding or GH bioactivity.
Structures of a number of cytokines, including G-CSF (Hill et al., 1993), GM-CSF (Diederichs et al., 1991; Walter et al., 1992), IL-2 (Bazan, 1992; McKay, 1992), IL-4 Redfield et al., 1991; Powers et al., 1992), and IL-5 (Milbum et al., 1993) have been determined by X-ray diffraction and NMR studies and show striking conservation with the GH structure, despite a lack of significant primary sequence homology. EPO is considered to be a member of this family based upon modeling and mutagenesis studies (Boissel et al., 1993; Wen et al., 1994). A large number of additional cytokines and growth factors including ciliary neurotrophic factor (CNTF), leukemia inhibitory factor (LIF), thrombopoietin (TPO), oncostatin M, macrophage colony stimulating factor (M-CSF), IL-3, IL-6, IL-7, IL-9, IL-12, IL-13, IL-15, and alpha, beta, omega, tau and gamma interferon belong to this family (reviewed in Mott and Campbell, 1995; Silvennoinen and Ihle 1996). All of the above cytokines and growth factors are now considered to comprise one large gene family, of which GH is the prototype.
In addition to sharing similar secondary and tertiary structures, members of this family share the property that they must oligomerize cell surface receptors to activate intracellular signaling pathways. Some GH family members, GH and EPO, bind a single type of receptor and cause it to form homodimers. Other family members, IL-2, L-4, and IL-6, bind more than one type of receptor and cause the receptors to form heterodimers or higher order aggregates (Davis et al., 1993; Paonessa et al., 1995; Mott and Campbell, 1995).
Mutagenesis studies have shown that, like GH, these other cytokines and growth factors contain multiple receptor binding sites, typically two, and bind their cognate receptors sequentially (Mott and Campbell, 1995; Matthews et al., 1996). Like GH, the primary receptor binding sites for these other family members occur primarily in the four alpha helices 1 5 and the A-B loop (reviewed in Mott and Campbell, 1995). The specific amino acids in the Shelical bundles that participate in receptor binding differ amongst the family members (Mott 0 and Campbell, 1995). Most of the cell surface receptors that interact with members of the GH supergene family are structurally related and comprise a second large multi-gene family O (Bazan, 1990; Mott and Campbell, 1995; Silvennoinen and Ihle 1996).
A general conclusion reached from mutational studies of various members of the GH supergene family is that the loops joining the alpha helices generally tend to not be involved in receptor binding. In particular the short B-C loop appears to be non-essential for receptor binding in most, if not all, family members. For this reason, the B-C loop is a preferred region for introducing cysteine substitutions in members of the GH supergene family. The A- B loop, the B-C loop, the C-D loop (and D-E loop of interferon/IL-10-like members of the GH superfamily) also are preferred sites for introducing cysteine mutations. Amino acids proximal to helix A and distal to the final helix also tend not to be involved in receptor binding and also are preferred sites for introducing cysteine substitutions. Certain members of the GH family, EPO, IL-2, IL-3, IL-4, IL-6, G-CSF, GM-CSF, TPO, IL-10, IL-12 IL-13, IL-15 and beta-interferon contain N-linked and O-linked sugars. The glycosylation sites in the proteins occur almost exclusively in the loop regions and not in the alpha helical bundles. Because the loop regions generally are not involved in receptor binding and because they are sites for the covalent attachment of sugar groups, they are preferred sites for introducing cysteine substitutions into the proteins. Amino acids that comprise the N- and 0linked glycosylation sites in the proteins are preferred sites for cysteine substitutions because these amino acids are surface-exposed, the natural protein can tolerate bulky sugar groups attached to the proteins at these sites and the glycosylation sites tend to be located away from the receptor binding sites.
Many additional members of the GH gene family are likely to be discovered in the future. New members of the GH supergene family can be identified through computer-aided secondary and tertiary structure analyses of the predicted protein sequences. Members of the GH supergene family will possess four or five amphipathic helices joined by non-helical amino acids (the loop regions). The proteins may contain a hydrophobic signal sequence at their N-terminus to promote secretion from the cell. Such later discovered members of the GH supergen family also are included within this invention.
The present invention provides "rules" for creating biologically active cysteine-added variants of members of the GH supergene family. These "rules" can be applied to any existing Sor future member of the GH supergene family. The cysteine-added variants will posses novel 8 properties not shared by the naturally occurring proteins. Most importantly, the cysteine added variants will possess the property that they can be covalently modified with cysteine- Sreactive polymers or other types of cysteine-reactive moieties to generate biologically active 0 proteins with improved properties such as increased in viv half-life, increased solubility and improved in vivo efficacy.
Specifically, the present invention provides biologically active cysteine variants of members of the GH supergene family by substituting cysteine residues for non-essential amino acids in the proteins. Preferably, the cysteine residues are substituted for amino acids that comprise the loop regions, for amino acids near the ends of the alpha helices and for amino acids proximal to the first amphipathic helix or distal to the final amphipathic helix of these proteins. Other preferred sites for adding cysteine residues are at the N-terminus or Cterminus of the proteins. Cysteine residues also can be introduced between two amino acids in the disclosed regions of the polypeptide chain. The present invention teaches that N- and O-linked glycosylation sites in the proteins are preferred sites for introducing cysteine substitutions either by substitution for amino acids that make up the sites or, in the case of Nlinked sites, introduction of cysteines therein. The glycosylation sites can be serine or threonine residues that are O-glycosylated or asparagine residues that are N-glycosylated.
N-
linked glycosylation sites have the general structure asparagine-X-serine or threonine (N-Xwhere X can be any amino acid. The asparagine residue, the amino acid in the X position and the serine/threonine residue of the N-linked glycosylation site are preferred sites for creating biologically active cysteine-added variants of these proteins. Amino acids immediately surrounding or adjacent to the O-linked and N-linked glycosylation sites (within about 10 residues on either side of the glycosylation site) are preferred sites for introducing cysteine-substitutions.
More generally, certain of the "rules" for identifying preferred sites for creating biologically active cysteine-added protein variants can be applied to any protein, not just proteins that are members of the GH supergene family. Specifically, preferred sites for c. creating biologically active cysteine variants of proteins (other than IL-2) are O-linked ID glycosylation sites. Amino acids immediately surrounding the O-linked glycosylation site (within about 10 residues on either side of the glycosylation site) also are preferred sites. Nlinked glycosylation sites, and the amino acid residues immediately adjacent on either side of the glycosylation site (within about 10 residues of the N-X-S/T site) also are preferred sites for creating cysteine added protein variants. Amino acids that can be replaced with cysteine without significant loss of biological activity also are preferred sites for creating cysteineadded protein variants. Such non-essential amino acids can be identified by performing cysteine-scanning mutagenesis on the target protein and measuring effects on biological activity. Cysteine-scanning mutagenesis entails adding or substituting cysteine residues for individual amino acids in the polypeptide chain and determining the effect of the cysteine substitution on biological activity. Cysteine scanning mutagenesis is similar to alaninescanning mutagenesis (Cunningham et al., 1992), except that target amino acids are individually replaced with cysteine rather than alanine residues.
Application of the "rules" to create cysteine-added variants and conjugates of protein antagonists also is contemplated. Excess production of cytokines and growth factors has been implicated in the pathology of many inflammatory conditions such as rheumatoid arthritis, asthma, allergies and wound scarring. Excess production of GH has been implicated as a cause of acromegaly. Certain growth factors and cytokines, GH and IL-6, have been implicated in proliferation of particular cancers. Many of the growth factors and cytokines implicated in inflammation and cancer are members of the GH supergene family. There is considerable interest in developing protein antagonists of these molecules to treat these diseases. One strategy involves engineering the cytokines and growth factors so that they can bind to, but not oligomerize receptors. This is accomplished by mutagenizing the second receptor binding site (site II) on the molecules. The resulting muteins are able to bind and occupy receptor sites but are incapable of activating intracellular signaling pathways. This strategy has been successfully applied to OH to make a GH antagonist (Cunningham et al., 1992). Similar strategies are being pursued to develop antagonists of other members of the GH supergene family such as IL-2 (Zurawski et al., 1990; Zurawski and Zurawski, 1992), IL- 4 (Kruse et al., 1992), IL-5 (Tavemier et al., 1995), GM-CSF (Hercus et al., 1994) and EPO (Matthews et al., 1996). Since the preferred sites for adding cysteine residues to members of the GH supergene family described here lie outside of the receptor binding sites in these 'Sb proteins, and thus removed from any sites used to create protein antagonists, the cysteineadded variants described herein could be used to generate long-acting versions of protein antagonists. As an example, Cunningham et al. (1992) developed an in vitro GH antagonist 1 5 by mutating a glycine residue (amino acid 120) to an arginine. This glycine residue is a critical component of the second receptor binding site in GH; when it is replaced with 8 arginine, GH cannot dimerize receptors. The glycine to arginine mutation at position 120 can be introduced into DNA sequences encoding the cysteine-added variants of GH contemplated herein to create a cysteine-added GH antagonist that can be conjugated with cysteine-reactive C o10 PEGs or other types of cysteine-reactive moieties. Similarly, amino acid changes in other proteins that turn the proteins from agonists to antagonists could be incorporated into DNA sequences encoding cysteine-added protein variants described herein. Considerable effort is being spent to identify amino acid changes that convert protein agonists to antagonists.
Hercus et al.(1994) reported that substituting arginine or lysine for glutamic acid at position 21 in the mature GM-CSF protein converts GM-CSF from an agonist to an antagonist.
Tavemier et a1.(1995) reported that substituting glutamine for glutamic acid at position 13 of mature IL-5 creates an IL-5 antagonist.
Experimental strategies similar to those described above can be used to create cysteine-added variants (both agonists and antagonists) of members of the GH supergene family derived from various animals. This is possible because the primary amino acid sequences and structures of cytokines and growth factors are largely conserved between human and animal species. For this reason, the "rules" disclosed herein for creating biologically active cysteine-added variants of members of the GH supergene family will be useful for creating biologically active cysteine-added variants of members of the GH supergene family of companion animals dogs, cats, horses) and commercial animal cow, sheep, pig) species. Conjugation of these cysteine-added variants with cysteinereactive PEGs will create long-acting versions of these proteins that will benefit the companion animal and commercial farm animal markets.
Proteins that are members of the GH supergene family (hematopoietic cytokines) are provided in Silvennoimem and Ihle (1996). Silvennoimem and Ihle (1996) also provide information about the structure and expression of these proteins. DNA sequences, encoded amino acids and in vitro and in ive bioassays for the proteins described herein are described in Aggarwal and Gutterman (1992; 1996), Aggarwal (1998), and Silvennoimem and Ihle _n (1996). Bioassays for the proteins also are provided in catalogues of various commercial suppliers of these proteins such as R&D Systems, Inc. and Endogen, Inc.
The following examples are provided to demonstrate how these "rules" can be used to create cysteine-added variants of OH, erythropoietin, alpha interferon, beta interferon, G- CSF, GM-CSF and other members of the GH supergene family. The examples are not 8 intended to be limiting, but only exemplary of specific embodiments of the invention.
Example 1 Cysteine-added variants of GH This example discloses certain amino acids in GH that are non-essential for biological activity and which, when mutated to cysteine residues, will not alter the normal disulfide binding pattern and overall conformation of the molecule. These amino acids are located at the N-terminal end of the A-B loop (amino acids 34-52 of the mature protein sequence; SEQ ID NO: 1; Martial et al 1979; Goeddel et al 1979), the B-C loop (amino acids 97-105 in the mature protein sequence), and the C-D loop (amino acids 130-153 in the mature protein sequence). Also identified as preferred sites for introducing cysteine residues are the first three or last three amino acids in the A, B, C and D helices and the amino acids proximal to helix A and distal to helix D.
DNA sequences encoding wild type GH can be amplified using the polymerase chain reaction technique from commercially available single-stranded cDNA prepared from human pituitaries (ClonTech, San Diego, CA) or assembled using overlapping oligonucleotides.
Specific mutations can be introduced into the GH sequence using a variety of procedures such as phage techniques (Kunkel et al 1987), PCR mutagenesis techniques (Innis et al 1990; White 1993) mutagenesis kits such as those sold by Stratagene ("Quick-Change Mutagenesis" kit, San Diego, CA) or Promega (Gene Editor Kit, Madison WI).
Cysteine substitutions can be introduced into any of the amino acids comprising the B-C loop, C-D loop and N-terminal end of the A-B loop or into the first three amino acids of the alpha helical regions that adjoin these regions. or in the region proximal to helix A or distal to helix D. Preferred sites for introduction of cysteine residues are: Fl, T3, P5, E33, A34, K38, E39, Q40, S43, Q46, N47, P48, Q49, T50, S51, S55, T60, A98, N99, S100, G104, A105, S106, E129, D130, G131, S132, P133, T135, G136, Q137, K140, Q141, T142, S144, K145, D147, T148, N149, S150, H151, N152, D153, S184, E186, G187, S188, and G190.
Cysteine residues also can be introduced at the beginning of the mature protein, proximal to the Fl amino acid, or following the last amino acid in the mature protein, following F191. If desirable, two or more such mutations can be readily combined in the same protein either by in vitro DNA recombination of cloned mutant genes and/or sequential construction of individual desired mutations.
i. Cloning the Gene for human Growth Hormone (GH) The human GH gene was amplified from human pituitary single-stranded cDNA (commercially available from CLONTECH, Inc., Palo Alto, CA) using the polymerase chain to reaction (PCR) technique and primers BBI and BB2. The sequence ofBBI is
GGGGGTCGACCATATGTTCCCAACCATTCCCTTATCCAG-
3 (SEQ ID NO: 24). The sequence of BB2 is GGGGGATCCTCACTAGAAGCCACAGCTGCCCTC 3' (SEQ ID NO: 25). Primer BB1 was designed to encode an initiator methionine preceding the first amino acid of mature GH, phenylalanine, and Sail and Ndel sites for cloning purposes. The reverse primer, BB2, contains a BamHI site for cloning purposes. The PCR 100 microliter reactions contained 20 pmoles of each oligonucleotide primer, IX PCR buffer (Perkin-Elmer buffer containing MgCl2), 200 micromolar concentration of each of the four nucleotides dA, dC, dG and dT, 2 ng of single-stranded cDNA, 2.5 units of Taq polymerase (Perkin-Elmer) and 2.5 units of Pfu polymerase (Stratagene, Inc). The PCR reaction conditions were 96 0
C
for 3 minutes, 35 cycles of(95 0 C, 1 minute; 63°C for 30 seconds; 72 0 C for 1 minute), followed by 10 minutes at 72 0 C. The thermocycler employed was the Amplitron II Thermal Cycler (Thermolyne). The approximate 600 bp PCR product was digested with Sail and BamHI, gel purified and cloned into similarly digested plasmid pUC19 (commercially available from New England BioLabs, Beverly, MA). The ligation mixture was transformed into E. coli strain DH5alpha and transformants selected on LB plates containing ampicillin.
Several colonies were grown overnight in LB media and plasmid DNA isolated using miniplasmid DNA isolation kits purchased from Qiagen, Inc (Valencia, CA). Clone LB6 was determined to have the correct DNA sequence.
For expression in E. cliclone LB6 was digested with NdeI and EcoRI, the approximate 600 bp fragment gel-purified, and cloned into plasmid pCYB 1 (commercially available from New England BioLabs, Beverly, MA) that had been digested with the same enzymes and phosphatased. The ligation mixture was transformed into E. coli DH5alpha and transformants selected on LB ampicillin plates. Plasmid DNA was isolated from several n- transformants and screened by digestion with NdeI and EcoRI. A correct clone was identified N and named pCYB1: wtGH (pBBT120). This plasmid was transformed into E. cp strains .JM109 or W3110 (available from New England BioLabs and the American Type Culture Collection).
2. Construction of STII-GH 9 Wild type GH clone LB6 (pUC19: wild type GH) was used as the template to 0 construct a GH clone containing theE. coli STII signal sequence (Picken et al. 1983).
SBecause of its length, the STII sequence was added in two sequential PCR reactions. The i C 10 first reaction used forward primer BB12 and reverse primer BBIO. BB0I has the sequence: 3' (SEQ ID NO: 28).
BB12 has the sequence: TTATCCAG-3' (SEQ ID NO: The PCR reactions were as described for amplifying wild type GH except that approximately 4 ng of plasmid LB6 was used as the template rather than single-stranded cDNA and the PCR conditions were 960C for 3 minutes, 30 cycles of (950C for 1 minute; 630C for 30 seconds; 72 0 C for 1 minute) followed by 72°C for 10 minutes. The approximate 630 bp PCR product was gel-purified using the Qiaex II Gel Extraction Kit (Qiagen, Inc), diluted 50-fold in water and 2 microliters used as template for the second PCR reaction. The second PCR reaction used reverse primer BB10 and forward primer BB 11. BB 11 has the sequence: TTTTCTCTATCG-3' (SEQ ID NO: 29).
Primer BB 11 contains Xbal and Ndel sites for cloning purposes. PCR conditions were as described for the first reaction. The approximate 660 bp PCR product was digested with Xbal and BamHL gel-purified and cloned into similarly cut plasmid pCDNA3. Invitrogen, Inc. Carlsbad, CA). Clone pCDNA3.1 (+)::stlI-GH(5C) or "5C" was determined to have the correct DNA sequence.
Clone "5C" was cleaved with NdeI and BamHI and cloned into similarly cut pBBT108 (a derivative of pUCl9 which lacks a Ps I site, this plasmid is described below). A clone with the correct insert was identified following digestion with these enzymes. This clone, designated pBBTI 11, was digested with Ndel and Sail, the 660 bp fragment containing n the stII-GH fusion gene, was gel-purified and cloned into the plasmid expression vector pCYB1 (New England BioLabs) that had been digested with the same enzymes and phosphatased. A recombinant plasmid containing the stII-GH insertion was identified by restriction endonuclease digestions. One such isolate was chosen for further studies and was designated pBBT114. This plasmid was transformed into E. coli strains JM109 or W3110 O (available from New England BioLabs and the American Type Culture Collection).
IN 3. Construction of ompA-GH SWild type GH clone LB6 (pUC19: wild type GH) was used as the template to Ci o10 construct a GH clone containing the E coli ompA signal sequence (Mowa et al 1980).
Because of its length, the ompA sequence was added in two sequential PCR reactions. The first reaction used forward primer BB7:
S'GCAGTGGCACTGCGGTTTCGCTACCGTAGCGCAGGCCTTCCCAACCATTCCC
TTATCCAG 3' (SEQ ID NO: 31), and reverse primer CGCGGATCCGATTAGAATCCACAGCTCCCCTC 3' (SEQ ID NO: 28).
The PCR reactions were as described for amplifying wild type OH except that approximately 4 ng of plasmid LB6 was used as the template rather than single-stranded cDNA and the PCR conditions were 96 0 C for 3 minutes, 30 cycles of (95°C for 1 minute; 63 0 C for 30 seconds; 72 0 C for 1 minute) followed by 72 0 C for 10 minutes. The approximate 630 bp PCR product was gel-purified using the Qiaex 11 Gel Extraction Kit (Qiagen, Inc), diluted 50-fold in water and 2 microliters used as template for the second PCR reaction. The second PCR reaction used reverse primer BBIO and forward PrimerBB6: GGTTTC 3' (SEQ ID NO: 32).
PCR conditions were as described for the first reaction. The approximate 660 bp PCR product was gel-purified, digested with Sal I and Bam HI and cloned into pUC19 (New England BioLabs) which was cut with al I and Bam HI or pCDNA3.1(+) (Invitrogen) which had been cut by Xho I and Barn HI Sal I and Xho I produce compatible single-stranded overhangs). When several clones were sequenced, it was discovered that all pUC19 clones contained errors in the region of the ompA sequence. Only one pCDNA3.1(+) clone was sequenced and it contained a sequence ambiguity in the ompA region. In order to generate a correct ompA-GH fusion gene segments of two sequenced clones which contained different errors separated by a convenient restriction site were IN recombined and cloned into the pUC19-derivative that lacks the Ps I site (see pBBT 08 described below). The resulting plasmid, termed pBBTI 12, carries the ompA-GH fusion gene cloned as an Nde I Bam HI fragment into these same sites in pBBTI08. This plasmid is designated pBBT1 12 and is used in PCR-based, site-specific mutagenesis of GH as described below.
I 4. Construction of Pst- pUC19 STo facilitate mutagenesis of the cloned GH gene for construction of selected cysteine substitution and insertion mutations a derivative of the plasmid pUC19 (New England BioLabs) lacking a Pst I site was constructed as follows. pUC19 plasmid DNA was digested with Pst I and subsequently treated at 75 deg. C with PFU DNA Polymerase (Stratagene) using the vendor-supplied reaction buffer supplemented with 200uM dNTPs. Under these conditions the polymerase will digest the 3' single-stranded overhang created by Pst I digestion but will not digest into the double-stranded region. The net result will be the deletion of the 4 single-stranded bases which comprise the middle four bases of the Pst I recognition site. The resulting molecule has double-stranded, "blunt", ends. Following these enzymatic reactions, the linear monomer was gel-purified using the Qiaex II Gel Extraction Kit (Qiagen, Inc). This purified DNA was treated with T4 DNA Ligase (New England BioLabs) according to the vendor protocols, digested with Ps 1, and used to transform E coli DHSalpha. Transformants were picked and analyzed by restriction digestion with Pst I and Bamr HI. One of the transformants which was not cleaved by EP I but was cleaved at the nearby Barn HI site was picked and designated pBBTIO8.
Construction ofGH muteins GH muteins were generally constructed using site-directed PCR-based mutagenesis as described in PCR Protocols: Current Methods and Applications edited by B. A. White, 1993 Humana Press, Inc., Totowa, NJ and PCR Protocols: A Guide to Methods and Applications edited by Innis, M. A. et al 1990 Academic Press Inc San Diego, CA. Typically PCR primer oligonucleotides are designed to incorporate nucleotide changes to the coding sequence of GH that result in substitution ofa cysteine residue for an amino acid at a specific position within the protein. Such mutagenic oligonucleotide primers can also be designed to incorporate an additional cysteine residue at the carboxy terminus or amino terminus of the coding sequence of GH. In this latter case one or more additional amino acid residues could n also be incorporated at the amino terminal and/or carboxy terminal to the added cysteine I residue if that were desirable. Moreover, oligonucleotides can be designed to incorporate cysteine residues as insertion mutations at specific positions within the GH coding sequence if that were desirable. Again, one or more additional amino acids could be inserted along with the cysteine residue and these amino acids could be positioned at the amino terminal and/or carboxy terminal to the cysteine residue.
IO The cysteine substitution mutation T135C was constructed as follows. The mutagenic reverse oligonucleotide BB28: 5'CTGCTTGAAGATCTGCCCACACCGGGGGCTGCCATC3' (SEQ ID NO: 33) was designed to change the codon ACT for threonine at amino acid residue 135 to a TGT codon encoding cysteine and to span the nearby Bgl II site. This oligonucleotide was used in PCR along with the forward oligonucleotide BB34 5'GTAGCGCAGGCCTTCCCAACCATT3' (SEQ ID NO: 34) which anneals to the junction region of the ompA-GH fusion gene and is not mutagenic. The PCR was performed in a reaction in IX PCR buffer (Perkin-Elmer buffer containing 1.5 mM MgClz), 200 micromolar concentration of each of the four nucleotides dA, dC, dG and dT, with each oligonucleotide primer present at 0.5 M, 5 pg ofpBBTl 12 (described above) as template and 1.25 units of Amplitac DNA Polymerase (Perkin-Elmer) and 0.125 units of PFU DNA Polymerase (Stratagene). Reactions were performed in a Robocycler Gradient 96 thermal cycler (Stratagene). The program used entailed: 95 deg C for 3 minutes followed by 25 cycles of deg C for 60 seconds, 45 deg C or 50 deg C or 55 deg C for 75 seconds, 72 deg C for seconds followed by a hold at 6 deg C. The PCR reactions were analyzed by agarose gel electrophoresis to identify annealing temperatures that gave significant product of the expected size; -430 bp. The 45-deg C reaction was "cleaned up" using the QIAquick PCR Purification Kit (Qiagen), digested with gl II and Pst I. The resulting 278 bp gl II- Pst I fragment, which includes the putative T135C mutation, was gel-purified and ligated into pBBTI 11 the pUCl9 derivative carrying the stII-GH fusion gene (described above) which had been digested with Bgl II and Pst I and gel-purified. Transformants from this ligation were initially screened by digestion with Bgl II and Pst I and subsequently one clone was sequenced to confirm the presence of the T135C mutation and the absence of any additional mutations that could potentially be introduced by the PCR reaction or by the synthetic Soligonucleotides. The sequenced clone was found to have the correct sequence.
Cl The substitution mutation S132C was constructed using the protocol described above for TI35C with the following differences: mutagenic reverse oligonucleotide BB29 5'CTGCTrGAAGATCTGCCCAGTCCGGGGGCAGCCATCTTC3' (SEQ ID NO: 35) was 'used instead of BB28 and the PCR reaction with annealing temperature of 50 deg C was used for cloning. One of two clones sequenced was found to have the correct sequence.
The substitution mutation T148C was constructed using an analogous protocol but (7 employing a different cloning strategy. The mutagenic forward oligonucleotide 5'GGGCAGATCTTCAAGCAGACCTACAGCAAGTTCGACTGCAACTCACACAAC 3 (SEQ ID NO: 36) was used in PCR with the non-mutagenic reverse primer BB33 5'CGCGGTACCCGGGATCCGATTAGAATCCACAGCT3' (SEQ ID NO: 37) which anneals to the most 3' end of the GH coding sequence and spans the Barn HI site immediately downstream. PCR was performed as described above with the exception that the annealing temperatures used were 46, 51 and 56 deg C. Following PCR and gel analysis as described above the 46 and 51 deg C reactions were pooled for cloning. These were digested with Barn HI and BgI I, gel-purified and cloned into pBBTI 11 which had been digested with Barn HI and Bgl EL, treated with Calf intestinal Alkaline Phosphatase (Promega) according to the vendor protocols, and gel-purified. Transformants from this ligation were analyzed by digestion with Barn HI and g..II to identify clones in which the 188 bp Barn HI- Bg II mutagenic PCR fragment was cloned in the proper orientation. Because Barn HI and BglII generate compatible ends, this cloning step is not orientation specific. Five of six clones tested were shown to be correctly oriented. One of these was sequenced and was shown to contain the desired T148C mutation. The sequence of the remainder of the 188 bp Barn HI B gL11 mutagenic PCR fragment in this clone was confirmed as correct.
The construction of the substitution mutation S 144C was identical to the construction of T148C with the following exceptions. Mutagenic forward oligonucleotide BB31 5'GGGCAGATCTrCAAGCAGACCTACTGCAAGTTCGAC3' (SEQ ID NO: 38) was used instead of BB30. Two of six clones tested were shown to be correctly oriented. One of these was sequenced and was shown to contain the desired S144C mutation. The sequence of the remainder of the 188 bp Bam HI- BglII mutagenic PCR fragment in this clone was confirmed as correct A mutation was also constructed that added a cysteine residue to the natural carboxy terminus of GH. The construction of this mutation, termed stpl92C, was similar to that of s T148C, but employed different oligonucleotide primers. The reverse mutagenic oligonucleotide BB32 5'CGCGGTACCGGATCCTTAGCAGAAGCCACAGCTGCCCTCCAC3' (SEQ ID NO: 39) which inserts a TGC codon for cysteine between the codon for the carboxy terminal phe residue of GH and the TAA translational stop codon and spans the nearby Bam HI site was to used along with BB34 5'GTAGCGCAGGCCTTCCCAACCATT3' (SEQ ID NO: 40) which is described above. Following PCR and gel analysis as described above, the 46 deg C reaction was used for cloning. Three of six clones tested were shown to be correctly oriented. One of these was sequenced and was shown to contain the desired stp192C mutation. The sequence of the remainder of the 188 bp Barn HI- BglnI mutagenic PCR fragment in this clone was confirmed as correct Analogous PCR mutagenesis procedures can be used to generate other cysteine mutations. The choice of sequences for mutagenic oligonucleotides will be dictated by the position where the desired cysteine residue is to be placed and the propinquity of useful restriction endonuclease sites. Generally, it is desirable to place the mutation, the mismatched segment near the middle of the oligonucleotide to enhance the annealing of the oligonucleotide to the template. Appropriate annealing temperatures for any oligonucleotide can be determined empirically, It is also desirable for the mutagenic oligonucleotide to span a unique restriction site so that the PCR product can be cleaved to generate a fragment that can be readily cloned into a suitable vector, one that can be used to express the mutein or provides convenient restriction sites for excising the mutated gene and readily cloning it into such an expression vector. Sometimes mutation sites and restriction sites are separated by distances that are greater than that which is desirable for synthesis of synthetic oligonucleotides: it is generally desirable to keep such oligonucleotides under 80 bases in length and lengths of 30-40 bases are more preferable.
In instances where this is not possible, genes targeted for mutagenesis could be reengineered or re-synthesized to incorporate restriction sites at appropriate positions.
Alternatively, variations of PCR mutagenesis protocols employed above, such as the so-called U "Megaprimer Method" (Barik, pp. 277-286 in Methods in Molecular Biology, Vol. Z PCR Protocols: Current Methods and Applications edited by B. A. White, 1993, Humana D Press, Inc., Totowa,NJ) or "Gene Splicing by Overlap Extension" (Horton, R. pp. 251- 261, in Methods in Molecular Biology, Vol. 15: PCR Protocols: Current Methods and Applications, edited by B. A. White, 1993, Humana Press, Inc., Totowa,NJ) can also be employed to construct such mutations.
C 6. Expression of GH in pCYBl Cr To express GH in E. coli pBBT 20 (GH gene with no leader sequence cloned into the Stac expression vector pCYBI) and pBBT114 (GH gene with stlI leader sequence cloned into C 10i the tac expression vector pCYB were transformed into E.coi strains JM109 and W3110.
The parental vector pCYBI was also transformed into JM109 and W3110.
These strains were given the following designations: BOB119: JM109 (pCYB1) BOB 130: W3110 (pCYBl) BOB129: JM109 BOB133: W3110 (pBBT120) BOB121: JM109 (pBBT 14) BOB132: W3110(pBBT114) For expression, strains were grown overnight at 37 C in Luria Broth (LB) (Sambrook, et al 1989) containing 100 ug/ml ampicillin. These saturated overnight cultures were diluted to 0.03 OD at A 6 oo in LB containing 100 gg/ml ampicillin and incubated at 37 C in shake flasks in rotary shaker, typically at 250-300 rpm. ODs were monitored and IPTG was added to a final concentration of 0.5 mM when culture ODs reached -0.25 typically between 0.3 and 0.4. Cultures were sampled typically at 1, 3, 5 and -16 h post-induction. The "~16 h" time points represented overnight incubation of the cultures and exact times varied from -15-20 h. Samples of induced and uninduced cultures were pelleted by centrifugation, resuspended in 1X sample buffer (50mM Tris-HC1 (pH 2% sodium lauryl sulfate, glycerol, 0.1% bromphenol blue) with the addition of 1% P-mercaptoethanol when desirable.
Samples were boiled for -10 minutes or heated to 95 C for 10 minutes. Samples were cooled to room temperature before being loaded onto SDS polyacrylamide gels or were stored at -200 C if not run immediately. Samples were run on precast 15% polyacrylamide "Ready Gels" (Bio-Rad, Hercules CA) using a Ready Gel Cell electrophoresis apparatus (Bio-Rad) according to the vendor protocols. Typically gels were run at 200 volts for -35-45 minutes.
Gels were stained with Coomassie Blue or were analyzed by Western Blot following electro- IND blotting. Coomassie staining of whole cell lysates from strains BOB 129, BOB 133, BOB 121 and BOB 132 showed a band of-22 kD that co-migrated with purified recombinant human GH standard purchased from Research Diagnostics Inc (Flanders, NJ). That band was _most prominent in induced cultures following overnight induction. However, a band was also observed at that molecular weight in uninduced cultures of these same strains and could also be observed with and without induction in the BOB 119 and BOB 130 control strains that carried the expression vector pCYBI lacking the GH gene. To clarify this observation, 17 10 Western Blot analyses were performed on whole cell lysates of induced cultures of strains BOB] 19, BOB] 30, BOB129, BOB133, BOB121, and BOB132. Western blots were performed with polyclonal rabbit anti-human GH antiserum purchased from United States Biological; catalogue G 9000-I 1 (Swampscott, MA) This primary antibody was used at a 1:5000 dilution and its binding was detected with goat anti-rabbit IgG Fc conjugated to Is alkaline phosphatase, (product 31341) purchased from Pierce (Rockford, IL). This secondary antibody was used at a 1:10,0000 dilution. Alkaline phosphatase activity was detected using the ImmunoPure® Fast Red TR/AS-MX Substrate Kit (Pierce, Rockford IL) according to the vendor protocols. The Western Blots clearly demonstrated presence of GH in lysates of induced cultures of BOBl29, BOB133, BOB121, and BOB132 at both 3 and 16 h post-induction. In the induced culture of control strains, BOBI 19 and BOB 130, no OH was detected by Western blot at 3 or 16 h post-induction time points.
In these preliminary experiments, the highest yields of GH were obtained from BOB132 W31 10(pBBT 114) in which the GH gene is fused downstream of the stll secretion signal sequence. This strain was tested further to determine if the GH protein was secreted to the periplasm as would be expected. An induced culture of BOB 132 was prepared as described above and subjected to osmotic shock according to the procedure of Koshland and Botstein (Cell 20 (1980) pp. 749-760). This procedure ruptures the outer membrane and releases the contents of the periplasm into the surrounding medium. Subsequent centrifugation separates the periplasmic contents, present in the supematant from the remainder of the cell-associated components. In this experiment, the bulk of the GH synthesized by BOB 132 was found to be localized to the periplasm. This result is consistent with the finding that the bulk of the total GH is also indistinguishable in size from the C purified GH standard, which indicated that the stII signal sequence had been removed. This Z is indicative of secretion. A larger scale (500 ml) culture of BOB132 was also induced, cultured overnight and subjected to osmotic shock according to the procedure described by Hsiung et al, 1986 (Bio/Technology 4, pp. 991-995). Gel analysis again demonstrated that the bulk of the GH produced was soluble, periplasmic, and indistinguishable in size from the GH standard. This material could also be quantitatively bound to, and eluted from, a Q- SSepharose column using conditions very similar to those described for recombinant human CN GH by Becker and Hsiung, 1986 (FEBS Lett 204 pp145-150).
S7, Cloning the human GH receptor CN 10 The human GH receptor was cloned by PCR using forward primer BB3 and reverse primer BB4. BB3 has the sequence: 5'-CCCCGGATCCGCCACCATGGATCTCTGGCAGCTGCTGTT-3' (SEQ ID NO: 26).
BB4 has the sequence: 5'CCCCGTCGACTCTAGAGCTATTAAATACGTAGCTCTTGGG-3' (SEQ ID NO: 27).
The template was single-stranded cDNA prepared from human liver (commercially available from CLONTECH Laboratories). Primers BB3 and BB4 contain BamHI and SaI restriction sites, respectively, for cloning purposes. The 100 jl PCR reactions contained 2.5 ng of the single-stranded cDNA and 20 picomolcs of each primer in IX PCR buffer (Perkin-Elmer buffer containing MgC12), 200 micromolar concentration of each of the four nucleotides dA, dC, dG and dT, 2.5 units ofTaq polymerase (Perkin-Elmer) and 2.5 units ofPfu polymerase (Stratagene, Inc). The PCR reaction conditions were: 96 0 C for 3 minutes, 35 cycles 1 minute; 58 0 C for 30 seconds; 72 0 C for 2 minutes), followed by 10 minutes at 72 0 C. The thermocycler employed was the Amplitron II Thermal Cycler (Thermolyne). The approximate 1.9 kb PCR product was digested with BamHI and Sail and ligated with similarly cut plasmid pUC19 (New England BioLabs). However, none of the transformants obtained from this ligation reaction contained the 1.9 kb PCR fragment. Leung et al (Nature 1987 330 pp537-543) also failed to obtain full-length cDNA clones of the human GH receptor in pUC19. Subsequently the PCR fragment was cloned into a low copy number vector, pACYC184 (New England BioLabs) at the Baw.HI and al sites in this vector. Such clones were obtained at reasonable frequencies but E coli strains carrying the cloned PCR fragment grew poorly, forming tiny and heterogeneous looking colonies, in the presence of chloramphenicol, which is used to select for maintenance ofpACYC184.
c The PCR fragment was simultaneously cloned into pCDNA3.1 (Invitrogen). The Z approximate 1.9 kb PCR product was digested with BamHI and Sall and ligated into the O BamHI and XhoI cloning sites of pCDNA3.1 Only infrequent transformants from this C' ligation contained the cloned GH receptor cDNA and all of those were found to contain deletions of segments of the receptor coding sequence. One of these clones was sequenced Sand found to contain a deletion of 135 bp within the GH receptor coding sequence: the Ssequence of the rest of the gene was in agreement with that reported by Leung et at (1987).
C1 8. Cloning the rabbit GH receptor 0The rabbit GH receptor was cloned by PCR using forward primer BB3 (described 10 above) and reverse primer BB36. BB36 has the sequence 3 (SEQ ID NO: 41) and contains XbaI and Sal I restriction sites for cloning purposes. Rabbit liver poly(A) mRNA was purchased from CLONTECH, Inc. and used as the substrate in first strand synthesis of single-stranded cDNA to produce template for PCR amplification. First strand synthesis of single-stranded cDNA was accomplished using a 1st Strand cDNA Synthesis Kit for RT-PCR (AMV) kit from Boehringer Mannheim Corp (Indianapolis, IN) according to the vendor protocols. Parallel first.strand cDNA syntheses were performed using random hexamers or BB36 as the primer. Subsequent PCR reactions with the products of the first strand syntheses as templates, were carried out with primers BB3 and BB36 according to the 1st Strand cDNA Synthesis Kit for RT-PCR (AMV) kit protocol and using 2.5 units of Amplitac DNA Polymerase (Perkin-Elmer) and 0.625 units ofPfu DNA Polymerase (Stratagene). The PCR reaction conditions were 96°C for 3 minutes, 35 cycles of (950C, 1 minute; 58 0 C for 30 seconds; 72°C for 2 minutes), followed by 10 minutes at 72 0 C. The thermocycler employed was the Amplitron II Thermal Cycler (Thermolyne). The expected 1.9 kb PCR product was observed in PCR reactions using random hexamer-primed or BB36 primed cDNA as template. The random hexamer-primed cDNA was used in subsequent cloning experiments. It was digested with Barn HI and Xbal and run out over a 1.2% agarose gel. This digest generates two fragments 365 bp and 1600 bp) because the rabbit GH receptor gene contains an internal Barn HI site. Both fragments were gel-purified. Initially the -1600 bp Barn H Xbal fragment was cloned into pCDNA3. which had been digested with these same two enzymes. These clones were readily obtained at reasonable frequencies and showed no evidence of deletions as determined by restriction digests and subsequent sequencing. To generate a full length clone, one of the plasmids containing the 1600 bp Ban HI Xba I fragment (pCDNA3.1(+):: rab-ghr-2A) was digested with Bamr HI, I treated with Calf Intestinal Alkaline Phosphatase (Promega) according to the vendor protocols, gel-purified and ligated with the gel purified -365 bp Bam HI fragment that contains the 5' portion of the rabbit GH receptor gene. Transfonnants from this ligation were picked and analyzed by restriction digestion and PCR to confirm the presence of the -365 bp C fragment and to determine its orientation relative to the distal segment of the rabbit GH receptor gene. Three out of four clones analyzed were found to contain the -365 bp fragment 0cloned in the correct orientation for reconstitution of the rabbit GH receptor gene. The lack So of complications in the cloning in E coli of the rabbit gene, in contrast to the human gene, is consistent with the results of Leung et al (1987) who also readily obtained full length cDNA clones for the rabbit GH receptor gene but were unable to clone a full length cDNA of the human gene in E coli. The rabbit GH receptor can be employed in assays with human GH as a ligand as it has been shown that the human GH binds the rabbit receptor with high affinity (Leung et al 1987). Plasmids containing the cloned rabbit GH receptor should be sequenced to identify a rabbit GH receptor cDNA with the correct sequence before use.
9.Construction of a human/rabbit Chimeric GH Receptor Gene As an alternative to the rabbit receptor, a chimeric receptor could be constructed which combines the extracellular domain of the human receptor with the transmembrane and cytoplasmic domains of the rabbit receptor. Such a chimeric receptor could be constructed by recombining the human and rabbit genes at the unique Nco I site that is present in each (Leung et al 1987). Such a recombinant, containing the human gene segment located 5' to, or "upstream" of the Nco I site and the rabbit gene segment 3' to, or "downstream" of the NC I site would encode a chimeric receptor of precisely the desired type, having the extracellular domain of the human receptor with the transmembrane and cytoplasmic domains of the rabbit receptor. This would allow analysis of the interaction ofGH, GH muteins, and PEGylated GH muteins with the natural receptor binding site but could avoid the necessity of cloning the full length human GH receptor in E coli.
The GH muteins can be expressed in a variety of expression systems such as bacteria, yeast or insect cells. Vectors for expressing GH muteins in these systems are available commercially from a number of suppliers such as Novagen, Inc. (pETI b for expression in E cli), New England Biolabs (pC4Bl for expression in E. coli Invitrogen (pVL1392, pVL1393, and pMELBAC for expression in insect cells using Baculovirus vectors, Pichia Svectors for expression in yeast cells, and pCDNA3 for expression in mammalian cells). GH IN has been successfully produced in E.col as a cytoplasmic protein and as a secreted, periplasmic, protein using the E. oli OmpA or STII signal sequences to promote secretion of 5 the protein into the periplasmic space (Chang et al., 1987; Hsiung et al., 1986). It is preferable that the GH muteins are expressed as secreted proteins so that they do not contain r n an N-terminal methionine residue, which is not present in the natural human protein. For expression in E. coli DNA sequences encoding GH or GH muteins can be cloned into E. coli 0expression vectors such as pET1 5b that uses the strong T7 promoter or pCYB 1 that uses the S 10 TAC promoter. Adding IPTG (isopropylthiogalactopyranoside, available form Sigma Chemical Company) to the growth media, can induce expression of the protein. Recombinant GH will be secreted into the periplasmic space from which it can be released by, and subsequently purified, following osmotic shock (Becker and Hsiung, 1986). The protein can be purified further using other chromatographic methods such as ion-exchange, hydrophobic interaction, size-exclusion and reversed phase chromatography, all of which are well known to those of skill in the art see Becker and Hsiung, 1986). Protein concentrations can be determined using commercially available protein assay kits such as those sold by BioRad Laboratories (Richmond, CA). If the GH proteins are insoluble when expressed in E. coli they can be refolded using procedures well known to those skilled in the art (see Cox et al., 1994 and World patent applications W09422466 and W09412219).
Alternatively, the proteins can be expressed in insect cells as secreted proteins. The expression plasmid can be modified to contain the OH signal sequence to promote secretion of the protein into the medium. The cDNAs can be cloned into commercially available vectors, pVL1392 from Invitrogen, Inc., and used to infect insect cells. The GH and GH muteins can be purified from conditioned media using conventional chromatography procedures. Antibodies to rhGH can be used in conjunction with Western blots to localize fractions containing the GH proteins during chromatography. Alternatively, fractions containing GH can be identified using ELISA assays.
The cysteine-added GH variants also can be expressed as intracellular or secreted proteins in eukaryotic cells such as yeast, insect cells or mammalian cells. Vectors for expressing the proteins and methods for performing such experiments are described in catalogues from various commercial supply companies such as Invitrogen, Inc., Stratagene, Inc. and ClonTech, Inc. The GH and GH muteins can be purified using conventional chromatography procedures.
IN Biological activity of the GH muteins can be measured using a cell line that proliferates in response to GH. Fuh et al. (1992) created a GH-responsive cell line by stably transforming a myeloid leukemia cell line, FDC-P1, with a chimeric receptor comprising the extracellular domain of the rabbit GH receptor fused to the mouse G-CSF receptor. This cell Sline proliferates in response to GH with a half maximal effective concentration (ECso) of picomolar. A similar cell line can be constructed using the published sequences of these receptors and standard molecular biology techniques (Fuh et al., 1992). Alternatively, the C 10 extracellular domain of the human GH receptor can be fused to the mouse G-CSF receptor using the published sequences of these receptors and standard molecular biology techniques.
Transformed cells expressing the chimeric receptor can be identified by flow cytometry using labeled GH, by the ability of transformed cells to bind radiolabeled GH, or by the ability of transformed cells to proliferate in response to added GH. Purified GH and GH muteins can be tested in cell proliferation assays using cells expressing the chimeric receptor to measure specific activities of the proteins. Cells can be plated in 96-well dishes with various concentrations of GH or GH muteins. After 18 h, cells are treated for 4 hours with 3
H-
thymidine and harvested for determination of incorporated radioactivity. The ECso can be determined for each mutein. Assays should be performed at least three times for each mutein using triplicate wells for each data point. GH muteins displaying similar optimal levels of stimulation and ECso values comparable to or greater than wild type GH are preferable.
GH muteins that retain in vitro activity can be PEGylated using a cysteine-reactive 8 kDa PEG-maleimide (or PEG-vinylsulfone) commercially available from Shearwater, Inc.
Generally, methods for PEGylating the proteins with these reagents will be similar to those described in world patent applications WO 9412219 and WO 9422466 and PCT application US95/06540, with minor modifications. The recombinant proteins must be partially reduced with dithiothreitol (DTT) in order to achieve optimal PEGylation of the free cysteine.
Although the free cysteine is not involved in a disulfide bond, it is relatively unreactive to cysteine-reactive PEGs unless this partial reduction step is performed. The amount of DTT required to partially reduce each mutein can be determined empirically, using a range of DTT concentrations. Typically, a 5-10 fold molar excess of DTT for 30 min at room temperature is sufficient Partial reduction can be detected by a slight shift in the elution profile of the protein from a reversed-phase column. Care must be taken not to over-reduce the protein and Sexpose additional cysteine residues. Over-reduction can be detected by reversed phase-HPLC ND (the protein will have a retention time similar to the fully reduced and denatured protein) and by the appearance of GH molecules containing two PEGs (detectable by a molecular weight change on SDS-PAGE). Wild type GH can serve as a control since it should not PEGylate under similar conditions. Excess DTT can be removed by size exclusion chromatography
C
using spin columns. The partially reduced protein can be reacted with various concentrations C' of PEG-maleimide (PEG: protein molar ratios of 1:1, 5:1,10:1 and 50:1) to determine the 0 optimum ratio of the two reagents. PEGylation of the protein can be monitored by a N i o molecular weight shift using sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE). The lowest amount of PEG that gives significant quantities of mono-pegylated product without giving di-pegylated product will be considered optimum (80% conversion to mono-pegylated product is considered good). Generally, mono-PEGylated protein can be purified from non-PEGylated protein and unreacted PEG by size-exclusion or ion exchange is chromatography. The purified PEGylated protein can be tested in the cell proliferation assay described above to determine its specific activity.
The above experiments will allow identification of amino acids in the B-C loop, C-D loop or N-terminal end of the A-B loop in GH that can be changed to cysteine residues, PEGylated and retain in vitro biological activity. These muteins can be tested in animal disease models well known in the art.
Experiments can be performed to confirm that the PEG molecule is attached to the protein at the proper site. This can be accomplished by proteolytic digestion of the protein, purification of the PEG peptide (which will have a large molecular weight) by size exclusion, ion exchange or reversed phase chromatography, followed by amino acid sequencing or mass spectroscopy. The PEG-coupled amino acid will appear as a blank in the amino acid sequencing run.
The pharmacokinetic properties of the PEG-GH proteins can be determined as follows, or as described in world patent application W09422466. Pairs of rats or mice can receive an intravenous bolus injection of the test proteins. Circulating levels of the proteins are measured over the course of 24 h by removing a small sample of blood from the animals at desired time points. Circulating levels of the test proteins can be quantitated using ELISA assays. Additional experiments can be performed using the subcutaneous route to administer
C
N the proteins. Similar experiments should be performed with the non-PEGylated protein to serve as a control. These experiments will reveal whether attachment of a PEG reagent to the IN protein alters its pharmacokinetic properties. Covalent modification of the protein with PEG should increase the protein's circulating half-life relative to the unPEGylated protein. Larger PEG molecules and/or attachment of multiple PEG molecules should lengthen the circulating \half-life longer than smaller PEG molecules.
tC PEG-GH proteins can be tested in rodent models of growth hormone deficiency (Cox 0 CN et al., 1994) and cachexia (Tomas et al., 1992; Read et al., 1992) to determine optimum Sdosing schedules and demonstrate efficacy. These studies can explore different size PEG C 100 molecules, 8 and 20 kDa, and dosing schedules to determine the optimum PEG size and dosing schedule. It is expected that the larger PEG molecule will increase the circulating half-life greater than the smaller PEG molecule and will require less frequent dosing.
However, large proteins potentially may have reduced volumes of distribution in ive; thus, it is possible a 20 kDa PEG attached to GH will limit bioavailability, reducing its efficacy.
Rodent models will allow determination of whether this is the case. Once the optimum dosing schedules and PEG sizes are determined, the efficacy of PEG-GH to GH can be compared in the animal models. While all PEG-GH proteins having GH activity are included in the invention, the preferred PEG-GH proteins are those that enhance growth equal or superior to GH, but which can be given less frequently. .PEG-GH should be more efficacious than GH when both are administered using the less frequent dosing schedules.
One GH deficiency model that can be used is a hypophysectomized rat. GH stimulates body weight gain and bone and cartilage growth in this model (Cox et al., 1994).
Hypophysectomized rats can be purchased from Charles River. Rats can be injected with GH, PEG-GH or placebo and weight gain measured daily over a 10-14 day period. At time of sacrifice, tibial epiphysis width can be determined as a measure of bone growth.
Experimental methods for performing these studies are described in Cox et al. (1994).
The efficacy of PEG-GH in rodent cachexia models can be tested in a similar manner.
Daily administration of dexamethasone, via osmotic pumps or subcutaneous injection, can be used to induce weight loss (Tomas et al., 1992; Read et al., 1992; PCT patent application US95/06540).
C' Example 2 Z Cysteine-added variants of Erythropoietin N. This example relates to cysteine-added variants of erythropoietin (EPO). EPO is the hormone primarily responsible for stimulating erythropoiesis or red blood cell formation.
EPO acts on immature red blood cell precursors to stimulate their further proliferation and differentiation into mature red blood cells. A commercial pharmaceutical version is available from Amgen, Inc. Human EPO is a 35-39 kDa glycoprotein secreted by the adult kidney.
The mature human protein contains 166 amino acids and is heavily glycosylated. The Ssequence of human EPO (SEQ ID NO: 2) is shown in Lin et al 1985 and Jacobs et al. 1985, io which are incorporated herein by reference. The primary sequence of EPO is highly conserved among species (greater than 80% identity; Wen et al., 1994). Sugar groups account for greater than 40% of the protein's mass. Human EPO contains three N-linked glycosylation sites and one O-linked glycosylation site. The N-linked glycosylation sites are conserved in different species whereas the O-linked glycosylation site is not. The extensive glycosylation of EPO has prevented the protein's crystallization, so the X-ray structure of the protein is not known. Human EPO contains four cysteine residues. The disulfide assignments are Cys7 to Cysl61 and Cys29 to Cys33. Cys33 is not conserved in mouse EPO, suggesting that the Cys29-Cys33 disulfide bond is not critical to mouse EPO's structure or function. This conclusion also seems to hold for human EPO (Boissel et al., 1993).
The amino acid sequence of EPO is consistent with the protein being a member of the GH supergene family and mutational studies support this view of EPO's structure (Boissel et al., 1993; Wen et al., 1994). A model of the three dimensional structure of EPO, modeled after the GH structure has been proposed (Boissel et al., 1993; Wen et al., 1994). Amino acids in EPO important for receptor binding have been identified through mutagenesis experiments and reside primarily in the N-terminal half of presumptive helix A and the Cterminal half of presumptive helix D (Boissel et al., 1993; Wen et al., 1994; Matthews et al., 1996). Only a single cell surface receptor for EPO has been identified (D'Andrea et al., 1989). It is believed that EPO dimerizes its receptor in much the same way that GH dimerizes its receptor (Matthew's et al., 1996).
Human EPO contains three sites for N-linked glycosylation (asparagine- 24,- 38 and 83) and one site for O-linked glycosylation (serine-12 6 The N-linked glycosylation sites are located in the A-B and B-C loops and the O-glycosylation site is located in the C-D loop.
c The N-linked glycosylation sites are conserved among species whereas the O-linked glycosylation site is absent in rodent EPO (Wen et al., 1993). A non-0-linked glycosylated I human variant containing methionine at position 126 has been described (US patent 4,703,008). The N-linked sugar groups are heavily branched and contain terminal sialic acid residues (Sasaki et al., 1987; Takeuchi et al., 1988). N-38 and N-83 contain the most highly Sbranched oligosaccharides (Sasaki et al., 1988).
SThe terminal sialic residues on EPO are critical for the protein's in viv function because removal of these residues by digestion eliminates iavivo activity (Fukada et al., O 1989; Spivak and Hogans, 1989). Loss of activity correlates with faster clearance of the o asialated protein from the body. The circulating half-life of the asialated protein in rats is less than ten minutes, in contrast to that of the sialated protein, which is approximately 2 hr (Fukada et al., 1989; Spivak and Hogans, 1989). Thus, in vivo activity of EPO directly correlates with its circulating half-life.
The role of N-linked sugars in EPO's biological activities haS been better defined by mutating, individually and in combination, the three asparagine residues comprising the Nlinked glycosylation sites. EPO muteins in which only a single N-linked glycosylation site was mutated, N24Q, N38Q and N83Q, were secreted from mammalian cells as efficiently as wild type EPO, indicating that N-linked glycosylation at all three sites is not required for protein secretion (N24Q indicates that the asparagine at position 24 is mutated to glutamine).
In contrast, EPO muteins in which two or more N-linked glycosylation sites were mutated were secreted less efficiently than wild type EPO from mammalian cells (Yamaguchi et al., 1991; Delorme et al., 1992). Mutagenesis studies found that each of the single N-linked glycosylation site muteins hadj i vitr biological activities equal to or greater than wild type EPO. Thus, it was concluded that none of the N-linked glycosylation sites is essential for secretion or in vitro biological activity of EPO. In fact, removal of one of the glycosylation sites seemed to improve biological activity (Yamaguchi et al., 1991).
The in vivo biological activity of N-linked glycosylation muteins was studied by two groups. Yamaguchi et al. (1991) concluded that the N24Q and N83Q muteins had in vi activities greater than wild type EPO, which correlated with their increased in vitro activities.
These authors found that the N38Q mutein had decreased in vivo activity, about 60% of wild type EPO. N38 is the most heavily branched of the three N-linked glycosylation sites (Sasaki et al., 1988). Delorme et al. (1992) reported that mutating any of the N-linked glycosylation c sites reduced in vivo biological activity by about 50%. Muteins in which two or more glycosylation sites were mutated had decreased in vivo activities in both studies.
The above studies indicate that some N-linked glycosylation is required for in vitro and in vivo activity of EPO. Individually, however, none of the three glycosylation sites is absolutely essential for activity. The N-linked sugars increase the apparent molecular weight of EPO and prolong its circulating half-life, which correlates with bioactivity. Natural EPO Cand EPO manufactured in mammalian cells have complex N-linked sugars containing 7 galactose and terminal sialic acid residues. The galactose residues are recognized by specific Sreceptors on hepatocytes and promote rapid clearance of EPO from the body unless the C 10 galactose residues are masked by the terminal sialic acid residues.
Mutagenesis studies concluded that 0-linked glycosylation is not required for in vitro or i vivo function of EPO (Delorme et al., 1992). This is in keeping with the observation that rodent EPO is not O-glycosylated and with the existence of a naturally occurring human EPO variant in which serine-126 is replaced by methionine, with a corresponding lack of 0linked glycosylation. Mutagenesis of serine-126 revealed that certain amino acid changes at this site (to valine, histidine or glutamic acid) yielded EPO muteins with biological activities similar to wild type EPO, whereas other amino acid changes (to alanine or glycine) resulted in EPO molecules with severely reduced activities (Delorme et al., 1992). The effect of changing serine-126 to cysteine was not studied. The in vivo bioactivity of S126V EPO was found to be similar to wild type EPO (Delorme et al., 1992).
The requirement for complex, N-linked carbohydrates containing terminal sialic acid residues for in viv activity of EPO has limited commercial manufacture of the protein to mammalian cells. The important functions of the sialated N-linked sugars are to prevent protein aggregation, increase protein stability and prolong the circulating half-life of the protein. The terminal sialic acid residues prolong EPO's circulating half-life by masking the underlying galactose residues, which are recognized by specific receptors on hepatocytes and promote clearance of the asialated protein. EPO can be produced in insect cells and is Nglycosylated and fully active in vitro; its activity in vivo has not been reported (Wojchowski et al., 1987).
This example provides for the design of cysteine-added EPO variants and their use in preparing conjugates using cysteine-reactive PEGs and other cysteine-reactive moieties.
Certain amino acids in EPO are non-essential for biological activity and can be mutated to cysteine residues without altering the normal disulfide binding pattern and overall Sconformation of the molecule. These amino acids are located in the A-B loop (amino acids 23-58 of the mature protein sequence), the B-C loop (amino acids 77-89 of the mature protein sequence), the C-D loop (amino acids 108-131 of the mature protein sequence), proximal to helix A (amino acids 1-8) and distal to helix D (amino acids 153-166 of the mature protein Ssequence). Also contemplated as preferred sites for adding cysteine residues are at the Nc terminus or C-terminus of the protein sequence. Preferred sites for cysteine substitutions are C the 0- linked glycosylation site (serine-126) and the amino acids comprising the three N- Slinked glycosylation sites (N24, 125, T26, N38, 139, T40, N83, S84, S85). Glycosylation sites Ci 0 are attractive sites for introducing cysteine substitutions and attaching PEG molecules to EPO because these sites are surface exposed; the natural protein can tolerate bulky sugar groups at these positions; the glycosylation sites are located in the putative loop regions and away from the receptor binding site (Wen et al., 1994); and mutagenesis studies indicate these sites (at least individually) are not essential for in viro or in vivo activity (Yamaguchi et al., 1991; Delorme et al., 1992). As discussed above, the local conformation of the region encompassing the O-glycosylation site region seems to be important for biological activity. Whether a cysteine substitution at position 126 affects biological activity has not been studied. The cysteine-29 to cysteine-33 disulfide bond is not necessary for biological activity of EPO because changing both residues to tyrosine simultaneously yielded a biologically active EPO protein (Boissel et al., 1993; Wen et al., 1994). A "free" cysteine can be created by changing either cysteine-29 or cysteine-33 to another amino acid. Preferred amino acid changes would be to serine or alanine. The remaining "free" cysteine (cysteine-29 or cysteine-33) would be a preferred site for covalently modifying the protein with cysteinereactive moieties.
Bill et al. (1995) individually substituted cysteine for N24, N38 and N83 and reported that the muteins had greatly reduced in vitro biological activities (less than 20% of wild type activity). Bill et al.(1995) expressed the EPO variants as fusion proteins (fused to glutathionine-S-transferase) in bacteria. One aspect of the present invention is to provide expression systems in which the N24C, N38C and N83C EPO variants will have in vitro biological activities more similar to wild type EPO.
US patent 4,703,008 contemplates naturally occurring variants of EPO as well as amino acid substitutions that are present in EPO proteins of mammals. Ovine EPO contains a C' cysteine residue at position 88 of the polypeptide chain. The inventor is unaware of any other Z naturally occurring human or animal cysteine variants of EPO in which the cysteine residue I occurs in the polypeptide regions disclosed herein as being useful for generating cysteinet C added EPO variants. Patent 4,703,008 specifically teaches away from cysteine-added EPO variants by suggesting that expression of EPO might be improved by deleting cysteine residues or substituting naturally occurring cysteine residues with serine or histidine residues.
SThe mature protein form of EPO can contain 165 or 166 amino acids because of post- -C translational removal of the C-terminal arginine. Asp-165 is the C-terminus of the 165 amino Sacid form and Agr-166 is the C-terminal amino acid of the 166 amino acid form. The C ito cysteine substitution and insertion mutations described herein can comprise either the 165 or 166 amino acid forms of mature EPO.
A cDNA encoding EPO can be cloned using the polymerase chain reaction (PCR) technique from the human HepG2 or Hep3B cell lines, which are known to express EPO when treated with hypoxia or cobalt chloride (Wen et al., 1993) and are available from the American Type Culture Collection (ATCC). Cysteine mutations can be introduced into the cDNA by standard phage, plasmid or PCR mutagenesis procedures as described for GH. As described above, the preferred sites for introduction of cysteine substitution mutations are in the A-B loop, the B-C loop, the C-D loop and the region proximal to helix A and distal to helix D. The most preferred sites in these regions are the N- and O-linked glycosylation sites: S126C; N24C; 125C, T26C; N38C; 139C, T40C; N83C, S 84C and S85C. Other preferred sites for cysteine substitution mutagenesis are in the A-B loop, the B-C loop and the C-D loop, amino acids surrounding the glycosylation sites and the region of the protein proximal to helix A and distal to helix D (Boissel et al., 1993; Wen et al., 1994). Other preferred sites for cysteine substitutions in these regions are: Al, P2, P3, R4, D8, S9, T27, G28, A30, E31, H32, S34, N36, D43, T44, K45, N47, A50, K52, E55, G57, Q58, G77, Q78, A79, Q86, W88, E89, T107, R110, All 1, G113, A114, Q115, K116, E117, Al 18, S120, P121, P122, D123, A124, A125, A127, A128, T132, K154, T157, G158, E159, A160, T163, G164, D 16 5 and R166. Cysteine residues also can be introduced proximal to the first amino acid of the mature protein, proximal to Al, or distal to the final amino acid in the mature protein, distal to D165 or R166. Other variants in which cys-29 or cys-33 have been replaced with other amino acids, preferably serine or alanine, also are provided.
C Wild type EPO and EPO muteins can be expressed using insect cells to determine Swhether the cysteine-added muteins are biologically active. DNAs encoding EPO/EPO \0 muteins can be cloned into the Baculovirus expression vector pVL1392 (available from Invitrogen, Inc. and Sigma Corporation (St. Louis, MO) and used to infect insect cells.
Recombinant Baculoviruses producing EPO can be identified by Western blots of infected insect cell conditioned media using polyclonal anti-human EPO antiserum (available from tC R&D Systems). The secreted EPO mutein proteins can be purified by conventional
O
CI chromatographic procedures well known to those of skill in the art. Protein concentrations Is0 Scan be determined using commercially available protein assay kits or ELISA assay kits C1 10 (available from R&D Systems and Bio-Rad Laboratories).
Purified EPO and EPO muteins can be tested in cell proliferation assays using EPOresponsive cell lines such as UT7-epo (Wen et al., 1994) or TFI (available from the ATCC) to measure specific activities of the proteins. Cells can be plated in 96-well microtiter plates with various concentrations of EPO. Assays should be performed in triplicate. After 1-3 days in culture, cell proliferation can be measured by 'H-thymidine incorporation, as described above for GH. The concentration of protein giving half-maximal stimulation (ECso) can be determined for each mutein. Assays should be performed at least three times for each mutein, with triplicate wells for each data point EC 5 o values can be used to compare the relative potencies of the muteins, Alternatively, cell proliferation in response to added EPO muteins can be analyzed using an MTT dye-exclusion assay (Komatsu et al.
1991). Proteins displaying similar optimal levels of stimulation and ECso values comparable to or greater than wild type EPO are preferable.
The above studies confirm identification of amino acid residues in EPO that can be changed to cysteine residues and retain biological activity. Muteins that retain activity can be PEGylated using a cysteine-reactive 8 kDa PEG-maleimide as described above for GH muteins. Wild type EPO should be used as a control since it should not react with the cysteine-reactive PEG under identical partial reduction conditions. The lowest amount of PEG that gives significant quantities of mono-PEGylated product without giving di- PEGylated product should be considered optimum. Mono-PEGylated protein can be purified from non-PEGylated protein and unreacted PEG by size-exclusion or ion exchange chromatography. The purified PEGylated proteins should be tested in the cell proliferation assay described above to determine their bioactivities.
C One or more of the PEGylated EPO muteins that retain in vitro bioactivity. are Scandidates for testing in animal disease models. PEGylation of the protein at the'proper amino IN acid can be determined as described for GH.
aInviv testing of PEGylated EPO muteins expressed using insect cells may require that they be re-engineered for expression in mammalian cells to ensure proper glycosylation.
O PEG-EPO candidates produced using insect cells can be tested in the animal models C described below to determine if they are active in viv and whether they are as active as PEG- NC EPO produced using mammalian cell expression systems. For expression in mammalian cells, O the EPO muteins can be subcloned into commercially available eukaryotic expression vectors o and used to stably transform Chinese Hamster Ovary (CHO) cells (available from the ATCC).
Sublines can be screened for EPO expression using ELISA assays. Sufficient quantities of the insect cell- and mammalian cell-produced EPO muteins can be prepared to compare their biological activities in animal anemia models.
In viva bioactivities of the EPO muteins can be tested using the artificial polycythemia or starved rodent models (Cotes and Bangham., 1961; Goldwasser and Gross., 1975). In the starved rodent model, rats are deprived of food on day one and treated with test samples on days two and three. On day four, rats receive an injection of radioactive iron-59.
Approximately 18h later, rats are anesthetized and blood samples drawn. The percent conversion of labeled iron into red blood cells is then determined. In the artificial polycythemia model, mice are maintained in a closed tank and exposed for several days to hypobaric air. The animals are then brought to normal air pressure. Red blood cell formation is suppressed for several days. On day four or six after return to normal air pressure, mice are injected with erythropoictin or saline. Mice receive one injection per day for one to two days.
One day later the animals receive an intravenous injection of labeled iron-59. The mice are euthanized 20 h later and the amount of labeled iron incorporated into red blood cells determined. EPO stimulates red blood cell formation in both models as measured by a dosedependent increase in labeled iron incorporated into red blood cells. In both models different dosing regimens and different times of injections can be studied to determine if PEG-EPO is biologically active and/or more potent and produces longer acting effects than natural EPO.
Example 3 Alpha Interferon c Alpha interferon is produced by leukocytes and has antiviral, anti-tumor and Z immunomodulatory effects. There are at least 20 distinct alpha interferon genes that encode IND proteins that share 70% or greater amino acid identity. Amino acid sequences of the known t C alpha interferon species are given in Blatt et al. (1996). A "consensus" interferon that incorporates the most common amino acids into a single polypeptide chain has been Sdescribed (Blatt et al., 1996). A hybrid alpha interferon protein may be produced by splicing m different parts of alpha interferon proteins into a single protein (Horisberger and Di Marco, 1995). Some alpha interferons contain N-linked glycosylation sites in the region proximal to Shelix A and near the B-C loop (Blatt et al. 1966). The alpha 2 interferon protein (SEQ ID NO: 3) contains four cysteine residues that form two disulfide bonds. The cysl-cys98 disulfide bond (cysl-cys99 in some alpha interferon species such as alpha 1; SEQ ID NO: 4) is not essential for activity. The alpha 2-interferon protein does not contain any N-linked glycosylation sites. The crystal structure of alpha interferon has been determined (Radhakrishnan et al., 1996).
This example provides cysteine added variants in the region proximal to the A helix, distal to the E helix, in the A-B loop, in the B-C loop, in the C-D loop and in the D-E loop.
Preferred sites for the introduction of cysteine residues in these regions of the alpha interferon-2 species are: D2, L3, P4, QS, T6, SS, Q20, R22, K23, S25, F27, S28, K31, D32, R33, D35, G37, F38, Q40, E41, E42, F43, 044, N45, Q46, F47, Q48, K49, A50, N65, S68, T69, K70, D71, S72, S73, A74, A75, D77, E78, T79, Y89, Q90, Q91, N93, D94, E96, A97, Q01, G102, G104, T106, E107, T108, P109, K112, El113, D14, S115, K131, E1 32 K1 3 3 K134, Y135, S136, A139, S152, S154, T155, N156, L157, Q158, E159, S160, L161, R162, S163, K164, E165. Variants in which cysteine residues are introduced proximal to the first amino acid of the mature protein, proximal to Cl, or distal to the final amino acid in the mature protein, distal to E165 are provided. Other variants in which cys-1 or cys-98 (cys- 99 in some alpha interferon species) have been replaced with other amino acids, preferably serine or alanine, also are provided. Other variants in which Cys-1 has been deleted (des Cys- 1) also are provided. The cysteine variants may be in the context of any naturally occurring or non-natural alpha interferon sequence, consensus interferon or interferon protein hybrids. Some naturally occurring alpha interferon species alpha interferon-1) contain a naturally occurring "free" cysteine. In such interferon species the naturally occurring free cysteine can be changed to another amino acid, preferably serine or alanine.
c This example also provides cysteine variants of other alpha interferon species, including consensus interferon, at equivalent sites in these proteins. The alignment of the O alpha interferon-2 species with other known alpha interferon species and consensus interferon
C
is given in Blatt et al. (1996). The crystal structure of alpha interferon -2 has been determined by Rhadhakrishnan et al. (1996). Lydon et al (1985) found that deletion of the first four amino acids from the N-terminus of alpha interferon did not affect biological activity.
SValenzuela et al (1985) found that substitution of Phe47 in alpha interferon-2 with Cys, Tyr or Ser did not alter biological activity of the protein. Cys-1 and Cys-98 have been changed o individually to glycine and serine, respectively, without altering biological activity of the C1 io protein (DeChiara et al, 1986).
DNA sequences encoding alpha interferon-2 can be amplified from human genomic DNA, since alpha interferon genes do not contain introns (Pestka et al., 1987). The DNA sequence of alpha interferon-2 is given in Goeddel et al. (1980). Alternatively, a cDNA for alpha interferon-2 can be isolated from human lymphoblastoid cell lines that are known to is express alpha interferon spontaneously or after exposure to viruses (Goeddell et al., 1980; Pickering et al., 1980). Many of these cell lines are available from the American Type Culture Collection (Rockville, MD). Specific mutations can be introduced into the alpha interferon sequence using plasmid-based site-directed mutagenesis kits Quick-Change Mutagenesis Kit, Stratagene, Inc.), phage mutagenesis strategies or employing PCR mutagenesis as described for GH.
Alpha interferon has been successfully produced in E. coli as an intracellular protein (Tamowski et al., 1986; Thatcher and Panayotatos, 1986). Similar procedures can be used to express alpha interferon muteins. Plasmids encoding alpha interferon or alpha interferon muteins can be cloned into an E.coli expression vector such as pETI5b (Novagene, Inc.) that uses the strong T7 promoter or pCYB (New England BioLabs, Beverly, MA) that uses the TAC promoter. Expression of the protein can be induced by adding IPTG to the growth media.
Recombinant alpha interferon expressed in E. coli is sometimes soluble and sometimes insoluble (Tarowski et al., 1986; Thatcher and Panayotatos, 1986). Insolubility appears to be related to the degree of overexpression of the protein. Insoluble alpha interferon proteins can be recovered as inclusion bodies and renatured to a fully active conformation following standard oxidative refolding protocols (Thatcher and Panayotatos, 1986; Cox et al., 1994). The alpha interferon proteins can be purified further using other chromatographic methods such as ion-exchange, hydrophobic interaction, size-exclusion and reversed phase resins (Thatcher and Panayotaos, 1986). Protein concentrations can be determined using commercially available protein assay kits (Bio-Rad Laboratories).
If col expression of alpha interferon muteins is not successful, one can express the proteins in insect cells as secreted proteins as described for GH. The proteins can be modified to contain the natural alpha interferon signal sequence (Goeddell et al., 1980) or the c'i honeybee mellitin signal sequence (Invitrogen, Inc.) to promote secretion of the proteins.
Alpha interferon and alpha interferon muteins can be purified from conditioned media using c'i 10 conventional chromatography procedures. Antibodies to alpha interferon can be used in conjunction with Western blots to localize fractions containing the alpha interferon proteins during chromatography. Alternatively, fractions containing alpha interferon proteins can be identified using ELISAs.
Bioactivities of alpha interferon and alpha interferon muteins can be measured using an in vitro viral plaque reduction assay (Ozes et al., 1992; Lewis, 1995). Human HeLa cells can be plated in 96-well plates and grown to near confluency at 370C. The cells are then washed and treated for 24 hour with different concentrations of each alpha interferon preparation. Controls should include no alpha interferon and wild type alpha interferon (commercially available from Endogen, Inc., Woburn, MA). A virus such as Vesicular stomatitis virus (VSV) or encephalomyocarditis virus (EMCV) is added to the plates and the plates incubated for a further 24-48 hours at 37 0 C. Additional controls should include samples without virus. When 90% or more of the cells have been killed in the virus-treated, no alpha interferon control wells (determined by visual inspection of the wells), the cell monolayer are stained with crystal violet and absorbance of the wells read using a microplate reader. Alternatively, the cell monolayers can be stained with the dye MTT (Lewis, 1995).
Samples should be analyzed in duplicate or triplicate. EC 50 values (the amount of protein required to inhibit the cytopathic effect of the virus by 50%) can be used to compare the relative potencies of the proteins. Wild type alpha interferon-2 protects cells from the cytopathic effects of VSV and EMCV and has a specific activity of approximately 2x10' units/mg in this assay (Ozes et al., 1992). Alpha interferon muteins displaying ECso values comparable to wild type Alpha Interferon are preferable.
C Alpha interferon muteins that retain activity can be PEGylated using procedures Ssimilar to those described for GH. Wild type alpha interferon-2 can serve as a control since it \should not PEGylate under similar conditions. The lowest amount of PEG that gives significant quantities of mono-pegylated product without giving di-pegylated product should be considered optimum. Mono-PEGylated protein can be purified from non-PEGylated protein and unreacted PEG by size-exclusion or ion exchange chromatography. The purified CPEGylated proteins can be tested in the viral plaque reduction bioassay described above to determine their bioactivities. PEGylated alpha interferon proteins with bioactivities O comparable to wild type alpha interferon are preferable. Mapping the PEG attachment site C" 10 and determination of pharmacokinetic data for the PEGylated protein can be performed as described for GH.
In vivo bioactivities of the PEG-alpha interferon muteins can be tested using tumor xenograft models in nude mice and viral infection models (Balkwill, 1986; Fish et al., 1986).
Since PEG-alpha interferon bioactivity may be species-specific, one should confirm activity of the PEGylated protein using appropriate animal cell lines in in vitro virus plaque reduction assays, similar to those described above. Next, one should explore the effects of different dosing regimens and different times of injections to determine if PEG-alpha interferon is more potent and produces longer lasting effects than non-PEGylated alpha interferon.
The novel alpha interferon-derived molecules of this example can be formulated and tested for activity essentially as set forth in Examples 1 and 2, substituting, however, the appropriate assays and other considerations known in the art related to the specific proteins of this example.
Example 4 Beta Interferon Beta interferon is produced by fibroblasts and exhibits antiviral, antitumor and immunomodulatory effects. The single-copy beta interferon gene encodes a preprotein that is cleaved to yield a mature protein of 166 amino acids (Taniguchi et al. 1980; SEQ ID NO: The protein contains three cysteines, one of which, cysteine-17, is "free", it does not participate in a disulfide bond. The protein contains one N-linked glycosylation site. The crystal structure of the protein has been determined (Karpusas et al. 1997)
I
CI This example provides cysteine-added variants at any of the three amino acids that Scomprise the N-linked glycosylation sites, N80C, E81C or T82C. This example also I provides cysteine-added variants in the region proximal to the A helix, distal to the E helix, in the A-B loop, in the B-C loop, in the C-D loop and in the D-E loop. Preferred sites for introduction of cysteine residues in these regions are: Ml, S2, Y3, N4, L5, Q23, N25, G26, SR27, E29, Y30, K33, D34, R35, N37, D39, E42, E43, K45, Q46, L47, Q48, Q49, Q51, K52, M E53, A68, F70, R71, Q72, D73, S74, S75, S76, T77, G78, E107, K108, El 09, D110, Fl l1, CN T112, R113, G114, K115, L116, A135, K136,E137, K138, S139,1157, N158, R159, L160, ST161, G162, Y163, L164, R165 and N166. Variants in which cysteine residues are introduced C 10 proximal to the first amino acid of the mature protein, proximal to Ml, or distal to the final amino acid in the mature protein, distal to N166 also are provided.
These variants are produced in the context of the natural protein sequence or a variant protein in which the naturally occurring "free" cysteine residue (cysteine-17) has been changed to another amino acid, preferably serine or alanine.
The novel beta interferon-derived molecules of this example can be formulated and tested for activity essentially as set forth in Examples I and 2, substituting, however, the appropriate assays and other considerations known in the art related to the specific proteins of this example.
Example Granulocyte Colony-Stimulating Factor (G-CSF) G-CSF is a pleuripotent cytokine that stimulates the proliferation, differentiation and function of granulocytes. The protein is produced by activated monocytes and macrophages.
The amino acid sequence of G-CSF (SEQ ID NO: 6) is given in Souza et al. (1986), Nagata et al. (1986a, b) and US patent 4,810,643 all incorporated herein by reference. The human protein is synthesized as a preprotein of 204 or 207 amino acids that is cleaved to yield mature proteins of 174 or 177 amino acids. The larger form has lower specific activity than the smaller form. The protein contains five cysteines, four of which are involved in disulfide bonds. Cysteine-17 is not involved in a disulfide bond. Substitution of cysteine-17 with serine yields a mutant G-CSF protein that is fully active (US patent 4,810,643). The protein is O-glycosylated at threonine-133 of the mature protein.
c1 This example provides a cysteine-added variant at threonine-133. This example Sprovides other cysteine-added variants in the region proximal to helix A, distal to helix D, in the A-B loop, B-C loop and C-D loop. Preferred sites for introduction ofcysteine N substitutions in these regions are: TI, P2, L3, G4, P5, A6, S7, S8, L9, P10, Q 11, S12, T38, K40, S53, G55, W58, A59, P60, S62, S63, P65, S66, Q67, A68, Q70, A72, Q90, A91, E93, O G94, S96, E98, G100, G125, M126, A127, A129, Q131, T133, Q134, G135, A136, A139, c A141, S142, A143, Q145, Q173 and P174. Variants in which cysteine residues are introduced C -proximal to the first amino acid of the mature protein, proximal to TI, or distal to the final amino acid in the mature protein, distal to P174 are provided. These variants are S0 provided in the context of the natural protein sequence or a variant protein in which the naturally occurring "free" cysteine residue (cysteine-17) has been changed to another amino acid, preferably serine or alanine.
A cDNA encoding human G-CSF can be purchased from R&D Systems (Minneapolis, MN) or amplified using PCR from mRNA isolated from human carcinoma cell lines such as 5637 and U87-MG known to express G-CSF constitutively (Park et al., 1989; Nagata, 1994).
These cell lines are available from the American Type Culture collection (Rockville, MD).
Specific mutations can be introduced into the G-CSF sequence using plasmid-based sitedirected mutagenesis kits Quick-Change Mutagenesis Kit, Stratagene, Inc.), phage mutagenesis methods or employing PCR mutagenesis as described for GH.
G-CSF has been successfully produced in E.coli as an intracellular protein Souza et al., 1986). One can employ similar procedures to express G-CSF and G-CSF muteins.
Plasmids encoding G-CSF or G-CSF muteins can be cloned into an E. coli expression vector such as pET15b (available from Novagen, Inc., Madison, WI) that uses the strong T7 promoter or pCYB1 (available from New England BioLabs, Beverly, MA) that uses the strong TAC promoter. Expression of the protein can be induced by adding IPTG to the growth media. Recombinant G-CSF expressed in E. coli is insoluble and can be recovered as inclusion bodies. The protein can be renatured to a fully active conformation following standard oxidative refolding protocols (Souza et al., 1986; Lu et al., 1992; Cox et al., 1994).
Similar procedures can be used to refold cysteine muteins. The proteins can be purified further using other chromatographic methods such as ion exchange, hydrophobic interaction, size-exclusion and reversed phase resins (Souza et al., 1986; Kuga et al., 1989; Lu et al., c 1992). Protein concentrations can be determined using commercially available protein assay Skits (Bio-Rad Laboratories).
I\ IfE. cl expression of G-CSF or G-CSF muteins is not successful, one can express G-CSF and G-CSF muteins in insect cells as secreted proteins as described for GH. The proteins can be modified to contain the natural G-CSF signal sequence (Souza et al., 1986; Nagata et al., 1986a; Nagata et al, 1986b) or the honeybee mellitin signal sequence C (Invitrogen, Inc., Carlsbad, CA) to promote secretion of the proteins. G-CSF and G-CSF NC muteins can be purified from conditioned media using conventional chromatography Sprocedures. Antibodies to G-CSF can be used in conjunction with Western blots to localize C- 10 fractions containing the G-CSF proteins during chromatography. Alternatively, fractions containing G-CSF proteins can be identified using ELISAs.
G-CSF muteins also can be expressed in mammalian cells as described for erythropoietin in Example 2.
Bioactivities of G-CSF and the G-CSF muteins can be measured using an in vitro cell proliferation assay. The mouse NFS-60 cell line and the human AML-193 cell line can be used to measure G-CSF bioactivity (Tsuchiya et al., 1986; Lange et al., 1987; Shirafuji et al., 1989). Both cell lines proliferate in response to human G-CSF. The AML-193 cell line is preferable since it is of human origin, which eliminates the possibility of a false conclusion resulting from species differences. The NFS60 cell lines proliferates in response to G-CSF with a half-maximal effective concentration (ECso) of 10-20 picomolar. Purified G-CSF and G-CSF muteins can be tested in cell proliferation assays using these cell lines to determine specific activities of the proteins, using published methods (Tsuchiya et al., 1986; Lange et al., 1987; Shirafuji et al, 1989). Cells can be plated in 96-well tissue culture dishes with different concentrations of G-CSF or G-CSF muteins. After 1-3 days at 37 0 C in a humidified tissue culture incubator, proliferation can be measured by 3 H-thymidine incorporation as described for GH. Assays should be performed at least three times for each mutein using triplicate wells for each data point. ECso values can be used to compare the relative potencies of the muteins. G-CSF muteins displaying similar optimal levels of stimulation and ECso values comparable to wild type G-CSF are preferable.
G-CSF muteins that retain activity can be PEGylated using procedures similar to those described for GH. Wild type G-CSF and ser-17 G-CSF can serve as controls since they should not PEGylate under similar conditions. The lowest amount of PEG that gives C' significant quantities of mono-PEGylated product without giving di-PEGylated product Sshould be considered optimum. Mono-PEGylated protein can be purified from non- IN PEGylated protein and unreacted PEG by size-exclusion or ion exchange chromatography.
The purified PEGylated proteins can be tested in the cell proliferation assay described above to determine their bioactivities.
The PEG site in the protein can be mapped using procedures similar to those Sdescribed for GH. Pharmacokinetic data for the PEGylated proteins can be obtained using
O
C procedures similar to those described for GH.
SInitial studies to demonstrate in vivo efficacy of PEG-G-CSF can be done in normal C 10 Sprague-Dawley rats, which can be purchased from Charles River. Groups of rats should receive single subcutaneous or intravenous injections of various doses of G-CSF, PEG-G- CSF or placebo. Animals should be sacrificed at daily intervals for up to a week for determination of neutrophil and total white blood cell counts in peripheral blood. Other blood cell types (platelets and red blood cells) can be measured to demonstrate cell specificity.
The efficacy of PEG-G-CSF can be tested in a rat neutropenia model. Neutropenia can be induced by treatment with cyclophosphamide, which is a commonly used chemotherapeutic agent that is myelosuppressive. O-CSF accelerates recovery of normal neutrophil levels in cyclophosphamide-treated animals (Kubota et al., 1990). Rats receive an injection of cyclophosphamide on day 0 to induce neutropenia. The animals are then be divided into different groups, which will receive subcutaneous injections of G-CSF, PEG-G- CSF or placebo. Neutrophil and total white blood cell counts in peripheral blood should be measured daily until they return to normal levels. Initially one should confirm that G-CSF accelerates recovery from neutropenia when injected daily. Next, one should explore the effects of different dosing regimens and different times of injections to determine if PEG-G- CSF is more potent and produces longer lasting effects than non-PEGylated G-CSF.
The novel GCSF-derived molecules of this example can be formulated and tested for activity essentially as set forth in Examples I and 2, substituting, however, the appropriate assays and other considerations known in the art related to the specific proteins of this example.
Example 6 Thrombopoietin (TPO) c-I Thrombopoietin stimulates the development of megakaryocyte precursors of platelets.
Z The amino acid sequence of TWO (SEQ ID NO: 7) is given in Bartley et aI.(]994), Foster et al.(1 994), de Sauvage et al.(I 994), each incorporated herein by reference.
The protein is synthesized as a 353 amino acid precursor protein that is cleaved to yield a mature protein of 332 amino acids. The N-tenninal 154 amino acids have homology with EPO and other members of the GH supergene family. The C-terminal 199 amino acids do not share homology with any other known proteins. The C-terminal region contains six N- CI linked glycosylation sites and multiple 0-linked glycosylation sites (Hoffman et al., 1996).
C) O-linked glycosylation sites also are found in the region proximal to the A helix, in the A-B cI 10 loop, and at the C-terminus of Helix C (Hoffmnan et al., 1996). A truncated TPO protein containing only residues 1- 195 of the mature protein is fully active in vitro (Bartley et al., 1994).
This example provides cysteine-added variants at any of the amino acids that comprise the N-inked glycosylation sites and the 0-linked glycosylation sites. This example also provides cysteine-added variants in the region proximal to the A helix, distal to the D helix, in the A-B loop, in the B-C loop, and in the C-D loop.
Preferred sites for introduction of cysteine residues are: SI, P2, A3, P4, P5, A6, T37, A43, D45, S47, G49, E50, K52, T53, Q54, E56, E57, T58, A76, A77, R78, G79, Q80, 082, T84, S87, S88, G109, TiIO1, Q1 11, P1 13, PI114, QlI15, GI116, RI117, TI 18, TI 19, A120, H121, K122, G146, G147, S148, T149, Al 55, TI158, TI59, A160, S163, T165, S166, TI170, NI176, RI177, T178, S179, 0180, El183, T184, N 185, F186, T187, A188, 51 89, A190, T192, T193, G194, S 195, N213, Q214, Tn15, S216, S218, N234, G235, T236, S244, T247, S254, S255, T257, S258, Tn60, S262, S272, S274, T-276, T280, T291, T294, S307, T3 10, T312, T-314, 53 15, N319, T320, S321, T323, S325, Q326, N327, L328, 5329, Q330, E331 and G332. Variants in which cysteine residues are introduced proximal to the first amino acid of the mature protein, proximal to SI1, or distal to the final amino acid in the mature protein, distal to G332 are provided. The cysteine-added variants are provided in the context of the natural human protein or a variant protein that is truncated between amino acids 147 and the C-terminus of the natural protein, G332. Variants in which cysteine residues are added distal to the final amino acid of a TPO protein that is truncated between amino acids 147 and 332 also are provided.
c1 The novel TPO-derived molecules of this example can be formulated and tested for S=activity essentially as set forth in Examples 1 and 2, substituting, however, the appropriate IN assays and other considerations known in the art related to the specific proteins of this example.
Example 7 Granulocyte-Macrophage Colony Stimulating Factor (GM-CSF) C GM-CSF stimulates the proliferation and differentiation of various hematopoietic Scells, including neutrophil, monocyte, eosinophil, erythroid, and megakaryocyte cell lineages.
io 1 The amino acid sequence of human GM-CSF (SEQ ID NO: 8) is given in Cantrell et al.
(1985) and Lee et al (1985) both incorporated herein by reference.
GM-CSF is produced as a 144 amino acid preprotein that is cleaved to yield a mature 127 amino acid protein. The mature protein has two sites for N-linked glycosylation. One site is located at the C-terminal end of Helix A; the second site is in the A-B loop.
This example provides cysteine-added variants at any of the amino acids that comprise the N-linked glycosylation sites, N27C, L28C, S29C, N37C, E38C and T39C. This example also provides cysteine-added variants in the region proximal to the A helix, distal to the D helix, in the A-B loop, in the B-C loop, and in the C-D loop. Preferred sites for introduction of cysteine substitutions in these regions are: Al, P2, A3, R4, SS5, P6, S7, PS, S9, TIO, Q11, R30, D31, T32, A33, A34, E35, E41, S44, E45, D48, Q50, E51, T53, Q64, R67, G68, S69, L70, T71, K72, K74, G75, T91, E93, T94, S95, A97, T98, T102, 1117, D120, E123, V125, Q126 and E127. Variants in which cysteine residues are introduced proximal to the first amino acid of the mature protein, proximal to Al, or distal to the final amino acid in the mature protein, distal to E127 are provided.
The novel GM-CSF-derived molecules of this example can be formulated and tested for activity essentially as set forth in Examples 1 and 2, substituting, however, the appropriate assays and other considerations known in the art related to the specific proteins of this example.
CN Example 8 S.IL-2 IL-2 is a T cell growth factor that is synthesized by activated T cells. The protein stimulates clonal expansion of activated T cells. Human IL-2 is synthesized as a 153 amino acid precursor that is cleaved to yield a 133 amino acid mature protein (Tadatsugu et al., S1983; Devos et al., 1983; SEQ ID NO: 9).
cThe amino acid sequence of IL-2 is set forth in (Tadatsugu et al. 1983; Devos et al.
-C 1983). The mature protein contains three cysteine residues, two of which form a disulfide bond. Cysteine-125 of the mature protein is not involved in a disulfide bond. Replacement Ci t10 of cysteine-125 with serine yields an IL-2 mutein with full biological activity (Wang et al., 1984). The protein is O-glycosylated at threonine-3 of the mature protein chain.
This example provides cysteine-added variants in the last four positions of the D helix, in the region distal to the D helix, in the A-B loop, in the B-C loop, and in the C-D loop. These variants are provided in the context of the natural protein sequence or a variant protein in which the naturally occurring "free" cysteine residue (cysteine-125) has been changed to another amino acid, preferably serine or alanine. Variants in which cysteine residues are introduced proximal to the first amino acid, Al, or distal to the final amino acid, T133, of the mature protein, also are provided.
The novel IL-2-derived molecules of this example can be formulated and tested for activity essentially as set forth in Examples 1 and 2, substituting, however, the appropriate assays and other considerations known in the art related to the specific proteins of this example.
Example 9 IL-3 IL-3 is produced by activated T cells and stimulates the proliferation and differentiation of pleuripotent hematopoietic stem cells. The amino acid sequence of human IL-3 (SEQ ID NO: 10) is given in Yang et al. (1986); Dorssers et al. (1987) and Otsuka et al.
(1988) all incorporated herein by reference. The protein contains two cysteine residues and two N-linked glycosylation sites. Two alleles have been described, resulting in isoforms having serine or proline at amino acid position 8 or the mature protein.
c This example provides cysteine-added variants at any of the amino acids that comprise Sthe N-linked glycosylation sites. This example also provides cysteine-added variants in the Sregion proximal to the A helix, distal to the D helix, in the A-B loop, in the B-C loop, and in the C-D loop. Variants in which cysteine residues are introduced proximal to the first amino acid or distal to the final amino acid in the mature protein, also are provided.
The novel L-3-derived molecules of this example can be formulated and tested for M activity essentially as set forth in Examples 1 and 2, substituting, however, the appropriate Ci assays and other considerations known in the art related to the specific proteins of this Sexample.
Example IL4 1L-4 is a pleiotropic cytokine that stimulates the proliferation and differentiation of monocytes and T and B cells. IL4 has been implicated in the process that leads to B cells secreting IgE, which is believed to play a role in asthma and atopy. The bioactivity of IL-4 is species specific. IL-4 is synthesized as a 153 amino acid precursor protein that is cleaved to yield a mature protein of 129 amino acids. The amino acid sequence of human IL-4 (SEQ ID NO:11) is given in Yokota et al. (1986) which is incorporated herein by reference. The protein contains six cysteine residues and two N-linked glycosylation sites. The glycosylation sites are located in the A-B and C-D loops.
This example provides cysteine-added variants at any of the amino acids that comprise the N-linked glycosylation sites. This example also provides cysteine-added variants in the region proximal to the A helix, distal to the D helix, in the A-B loop, in the B-C loop, and in the C-D loop. Variants in which cysteine residues are introduced proximal to the first amino acid, H1, or distal to the final amino acid, S129, of the mature protein are provided.
The novel L-4-derived molecules of this example can be formulated and tested for activity essentially as set forth in Examples 1 and 2, substituting, however, the appropriate assays and other considerations known in the art related to the specific proteins of this example.
Example 11 is a differentiation and activation factor for eosinophils. The amino acid Ssequence of human IL-5 (SEQ ID NO: 12) is given in Yokota et al. (1987) which is incorporated herein by reference. The mature protein contains 115 amino acids and exists in solution as a disulfide-linked homodimer. The protein contains both O-linked and N-linked glycosylation sites.
This example provides cysteine-added variants in the region proximal to the A helix, c distal to the D helix, in the A-B loop, in the B-C loop, and in the C-D loop. Variants in which cysteine residues are added proximal to the first amino acid or distal to the final amino acid in Sthe mature protein are provided.
S 10 The novel IL-5-derived molecules of this example can be formulated and tested for activity essentially as set forth in Examples 1 and 2, substituting, however, the appropriate assays and other considerations known in the art related to the specific proteins of this example.
Example 12 IL-6 IL-6 stimulates the proliferation and differentiation of many cell types. The amino acid sequence of human IL-6 (SEQ ID NO: 13) is given in Hirano et al. (1986) which is incorporated herein by reference. IL-6 is synthesized as a 212 amino acid preprotein that is cleaved to generate a 184 amino acid mature protein. The mature protein contains two sites for N-linked glycosylation and one site for O-glycosylation, at T137, T138, T142 or T143.
This example provides cysteine-added variants at any of the amino acids that comprise the N-linked glycosylation sites and at the O-linked glycosylation site. Also provided are cysteine-added variants in the region proximal to the A helix, distal to the D helix, in the A-B loop, in the B-C loop, and in the C-D loop. Variants in which cysteine residues are added proximal to the first amino acid or distal to the final amino acid of the mature protein also are provided.
The novel IL-6-derived molecules of this example can be formulated and tested for activity essentially as set forth in Examples 1 and 2, substituting, however, the appropriate assays and other considerations known in the art related to the specific proteins of this example.
C Example 13 IIIL-7 s IL-7 stimulates proliferation of immature B cells and acts on mature T cells. The amino acid sequence of human IL-7 (SEQ ID NO: 14) is given in Goodwin et al. (1989) which is incorporated herein by reference. The protein is synthesized as a 177 amino acid 0preprotein that is cleaved to yield a 152 amino acid mature protein that contains three sites for aC N-linked glycosylation.
O
C, This example provides cysteine-added variants at any of the amino acids that comprise Sthe N-linked glycosylation sites. This example also provides cysteine-added variants in the C 10 region proximal to the A helix, distal to the D helix, in the A-B loop, in the B-C loop, and in the C-D loop.
The novel L-7-derived molecules of this example can be formulated and tested for activity essentially as set forth in Examples 1 and 2, substituting, however, the appropriate assays and other considerations known in the art related to the specific proteins of this example. Variants in which cysteine residues are added proximal to the first amino acid or distal to the final amino acid of the mature protein also are provided.
Example 14 IL-9 IL-9 is a pleiotropic cytokine that acts on many cell types in the lymphoid, myeloid and mast cell lineages. IL-9 stimulates the proliferation of activated T cells and cytotoxic T lymphocytes, stimulates proliferation of mast cell precursors and synergizes with erythropoietin in stimulating immature red blood cell precursors. The amino acid sequence of human IL-9 (SEQ ID NO: 15) is given in Yang et al. (1989) which is incorporated herein by reference. IL-9 is synthesized as a precursor protein of 144 amino acids that is cleaved to yield a mature protein of 126 amino acids. The protein contains four potential N-linked glycosylation sites.
This example provides cysteine-added variants at any of the three amino acids that comprise the N-linked glycosylation sites. This example also provides cysteine-added variants in the region proximal to the A helix, distal to the D helix, in the A-B loop, in the B-C loop, and in the C-D loop. Variants in which cysteine residues are added proximal to the first amino acid or distal to the final amino acid of the mature protein also are provided.
O
C1 The novel IL-9-derived molecules of this example can be formulated and tested for Sactivity essentially as set forth in Examples 1 and 2, substituting, however, the appropriate IN assays and other considerations known in the art related to the specific proteins of this example.
Example SThe amino acid sequence of human IL-10 (SEQ ID NO: 16) is given in Vieira et al.
(1991) which is incorporated herein by reference. IL-10 is synthesized as a 178 amino acid C 10 precursor protein that is cleaved to yield a mature protein of 160 amino acids. IL-10 can function to activate or suppress the immune system.. The protein shares structural homology with the interferons, it contains five amphipathic helices. The protein contains one Nlinked glycosylation site.
This example provides cysteine-added variants at any of the three amino acids comprising the N-linked glycosylation site. This example also provides cysteine-added variants in the region proximal to the A helix, distal to the E helix, in the A-B loop, in the B- C loop, in the C-D loop and in the D-E loop. Variants in which cysteine residues are added proximal to the first amino acid or distal to the final amino acid of the mature protein also are provided.
The novel IL-10-derived molecules of this example can be formulated and tested for activity essentially as set forth in Examples 1 and 2, substituting, however, the appropriate assays and other considerations known in the art related to the specific proteins of this example.
Example 16 IL-11 IL- 1 is a pleiotropic cytokine that stimulates hematopoiesis, lymphopoeisis and acute phase responses. IL-11 shares many biological effects with IL-6. The amino acid sequence of human IL- 1 (SEQ ID NO: 17) is given in Kawashima et al. (1991) and Paul et al. (1990) both incorporated herein by reference. IL-11 is synthesized as a precursor protein of 199 amino acids that is cleaved to yield a mature protein of 178 amino acids. There are no Nlinked glycosylation sites in the protein.
C1 This example provides cysteine-added variants in the region proximal to the A helix, Sdistal to the D helix, in the A-B loop, in the B-C loop, and in the C-D loop. Variants in which IN cysteine residues are added proximal to the first amino acid or distal to the final amino acid of c the mature protein also are provided.
5 The novel IL-11 -derived molecules of this example can be formulated and tested for ractivity essentially as set forth in Examples I and 2, substituting, however, the appropriate Ccr assays and other considerations known in the art related to the specific proteins of this example.
C- ;o Example 17 IL-12 IL-12 stimulates proliferation and differentiation of NK cells and cytotoxic T lymphocytes. IL-12 exists as a heterodimer of a p35 subunit and a p40 subunit. The subunit is a member of the GH supergene family. The amino acid sequence of the subunit (SEQ ID NO: 18) is given in Gubler et al. (1991) and Wolf et al. (1991) both incorporated herein by reference, p35 is synthesized as a precursor protein of 197 amino acids and is cleaved to yield a mature protein of 175 amino acids. The protein contains 7 cysteine residues and three potential N-linked glycosylation sites.
This example provides cysteine-added variants at any of the three amino acids that comprise the three N-linked glycosylation sites. This example also provides cysteine-added variants in the region proximal to the A helix, distal to the D helix, in the A-B loop, in the B- C loop, and in the C-D loop. These variants are provided in the context of the natural protein sequence or a variant protein in which the naturally occurring "free" cysteine residue has been changed to another amino acid, preferably serine or alanine. Variants in which cysteine residues are added proximal to the first amino acid or distal to the final amino acid of the mature protein also are provided.
The novel IL-12 p35-derived molecules of this example can be formulated and tested for activity essentially as set forth in Examples 1 and 2, substituting, however, the appropriate assays and other considerations known in the art related to the specific proteins of this example.
Example 18 Z IL-13 IL-13 shares many biological properties with IL-4. The amino acid sequence of human IL-13 (SEQ ID NO: 19) is given in McKenzie et al. (1993) and Minty et al. (1993) both incorporated herein by reference. The protein is synthesized as a 132 amino acid precursor protein that is cleaved to yield a mature protein of 112 amino acids. The mature Cc protein contains 5 cysteine residues and multiple N-linked glycosylation sites. A variant in
O
C which glutamine at position 78 is deleted due to alternative mRNA splicing has been 0described (McKenzie et al 1993) C 10 This example provides cysteine-added variants at any of the three amino acids comprising the N-linked glycosylation sites. This example also provides cysteine-added variants in the region proximal to the A helix, distal to the D helix, in the A-B loop, in the B- C loop, and in the C-D loop. These variants are provided in the context of the natural protein sequence or a variant sequence in which the pre-existing "free" cysteine has been changed to another amino acid, preferably to alanine or serine. Variants in which cysteine residues are added proximal to the first amino acid or distal to the final amino acid of the mature protein also are provided.
The novel IL-13-derived molecules of this example can be formulated and tested for activity essentially as set forth in Examples 1 and 2, substituting, however, the appropriate assays and other considerations known in the art related to the specific proteins of this example.
Example 19 IL-15 stimulates the proliferation and differentiation of T cells, NK cells, LAK cells and Tumor Infiltrating Lymphocytes. IL-15 may be useful for treating cancer and viral infections. The sequence of IL-15 (SEQ IDNO: 20) is given in Anderson et al. (1995) which is incorporated herein by reference. IL-15 contains two N-linked glycosylation sites, which are located in the C-D loop and C-terminal end of the D helix. IL-15 encodes a 162 amino acid preprotein that is cleaved to generate a mature 114 amino acid protein.
This example provides cysteine-added variants at any of the three amino acids comprising the N-linked glycosylation sites in the C-D loop or C-terminal end of the D helix.
This example also provides cysteine-added variants proximal to helix A, in the A-B loop, the B-C loop, the C-D loop or distal to helix D. Variants in which cysteine residues are added proximal to the first amino acid or distal to the final amino acid of the mature protein also are provided.
The novel IL-15-derived molecules of this example can be formulated and tested for activity essentially as set forth in Examples 1 and 2, substituting, however, the appropriate assays and other considerations known in the art related to the specific proteins of this example.
Example Macrophage Colony Stimulating Factor (M-CSF): M-CSF regulates the growth, differentiation and function ofmonocytes. The protein is a disulfide-linked homodimer. Multiple molecular weight species of M-CSF, which arise from differential mRNA splicing, have been described. The amino acid sequence of human M-CSF and its various processed forms are given in Kawasaki et al (1985), Wong et al.
(1987) and Cerretti et al (1988) which are incorporated herein by reference. Cysteine-added variants can be produced following the general teachings of this application and in accordance with the examples set forth herein.
The novel MCSF-derived molecules of this example can be formulated and tested for activity essentially as set forth in Examples 1 and 2, substituting, however, the appropriate assays and other considerations known in the art related to the specific proteins of this example.
Example 21 Oncostatin M: Oncostatin M is a multifunctional cytokine that affects the growth and differentiation of many cell types. The amino acid sequence of human oncostatin M (SEQ ID NO: 21) is given in Malik et al. (1989) which is incorporated herein by reference. Oncostatin M is produced by activated monocytes and T lymphocytes. Oncostatin M is synthesized as a 252 amino acid preprotein that is cleaved sequentially to yield a 227 amino acid protein and then a 196 amino acid protein (Linsley et al., 1990). The mature protein contains O-linked C'7 glycosylation sites and two N-linked glycosylation sites. The protein is O-glycosylated at T160, T162 and S165. The mature protein contains five cysteine residues.
C This example provides cysteine-added variants at either of the three amino acids C' comprising the N-linked glycosylation sites or at the amino acids that comprise the O-linked s glycosylation sites. This example also provides cysteine-added variants proximal to helix A, 0in the A-B loop, in the B-C loop, in the C-D loop or distal to helix D. These variants are Sprovided in the context of the natural protein sequence or a variant sequence in which the pre- C existing "free" cysteine has been changed to another amino acid, preferably to alanine or 0serine. Variants in which cysteine residues are added proximal to the first amino acid or distal C lo to the final amino acid of the mature protein also are provided.
The novel Oncostatin M-derived molecules of this example can be formulated and tested for activity essentially as set forth in Examples 1 and 2, substituting, however, the appropriate assays and other considerations known in the art related to the specific proteins of this example.
Example 22 Ciliary Neurotrophic Factor (CNTF): The amino acid sequence of human CNTF (SEQ ID NO: 22) is given in Lam et al.
(1991) which is incorporated herein by reference. CNTF is a 200 amino acid protein that contains no glycosylation sites or signal sequence for secretion. The protein contains one cysteine residue. CNTF functions as a survival factor for nerve cells.
This example provides cysteine-added variants proximal to helix A, in the A-B loop, the B-C loop, the C-D loop or distal to helix D. These variants are provided in the context of the natural protein sequence or a variant sequence in which the pre-existing "free" cysteine has been changed to another amino acid, preferably to alanine or serine. Variants in which cysteine residues are added proximal to the first amino acid or distal to the final amino acid of the mature protein also are provided.
The novel CNTF-derived molecules of this example can be formulated and tested for activity essentially as set forth in Examples 1 and 2, substituting, however, the appropriate assays and other considerations known in the art related to the specific proteins of this example.
cExample 23 ZLeukemia Inhibitory Factor (LIF) IThe amino acid sequence of LIF (SEQ ID NO: 23) is given in Moreau et al. (1988) and Gough et al. (1988) both incorporated herein by reference. The human gene encodes a 202 amino acid precursor that is cleaved to yield a mature protein of 180 amino acids. The \protein contains six cysteine residues, all of which participate in disulfide bonds. The protein Mr3 contains multiple 0- and N-linked glycosylation sites. The crystal structure of the protein cwas determined by Robinson et al. (1994). The protein affects the growth and differentiation of many cell types.
ci i0 This example provides cysteine-added variants at any of the three amino acids comprising the N-linked glycosylation sites or the O-linked glycosylation site. Also provided are cysteine-added variants proximal to helix A, in the A-B loop, the B-C loop, the C-D loop or distal to helix D. Variants in which cysteine residues are added proximal to the first amino acid or distal to the final amino acid of the mature protein also are provided.
The novel LIF-derived molecules of this example can be formulated and tested for activity essentially as set forth in Examples I and 2, substituting, however, the appropriate assays and other considerations known in the art related to the specific proteins of this example.
All of the documents cited herein are incorporated herein by reference.
The protein analogues disclosed herein can be used for the known therapeutic uses of the native proteins in essentially the same forms and doses all well known in the art.
While the exemplary preferred embodiments of the present invention are described herein with particularity, those having ordinary skill in the art will recognize changes, modifications, additions, and applications other than those specifically described herein, and may adapt the preferred embodiments and methods without departing from the spirit of this invention.
References: Abdel-Meguid, Shieh, Smith Dayringer, Violand, B.N. and Bentle, L.A. (1987) Proc. Natl. Acad. Sci. USA 84: 6434-6437.
I
c-i Abuchowski, Kazo, Verhoest, van Es, Kafkewitz, Nucci, Viau, Z A.T. and Davis, F.F. (1984) Cancer Biochem. Biophys. 7: 175-186.
Aggarwal, B.B. (1998) Human Cytokines. Handbook for Basic and Clinical Research.
CI Volume 111. Blackwell Science, Maiden, MA.
Aggarwal, B.B. and Gutterman, J.U. (1992) Human Cytokines. Handbook for Basic and Clinical Research. Volume 1. Blackwell Scientific Publications, Cambridge, MA.
Aggarwal, B.B. and Gutterman, J.U. (1996) Human Cytokines. Handbook for Basic and Clinical Research. Volume UI. Blackwell Science, Cambridge, M.A.
INO Anderson, Johnson, Glaccum, Copeland, Gilbert, DJ., Jenkins, N.A., Valentine, Kirstein, Shapiro, Morris, Grabstein, K. and cosinan, D.
(1995) Genomics 25: 701-706.
Balkwill, F.R (1986) Methods Enzymology 119: 649-657.
Bartley, Bogenberger, J. et al., (1994) Cell 77: 1117-1124.
Bazan, F. (1991) Immunology Today 11: 350-354.
Ba zan, J.F. (1992) Science 257: 410-411.
Becker, G.W. and Hsiung, H. M. (1986) FEBS Letters 204: 145-150O.
Bill, Winter, McHale, Hodges, Elder, Caley, Flitsch, S.L., Bicknell, R. Lappin, T.R.J. (1995) Biochem. Biophys. Acta 1261: 35-43.
Bittorf, Jaster, Brock, J. (1993) FEBS Letters 336: 133-136.
Blatt, Davis, Klein, S.B. and Taylor, M.W. (1996) Journal of Interferon and cytokine Research 16: 489-499.
Boissel, Lee, Presnell, Cohen, F.E. and Bunn, H.F. (1993) J. Biol. Chem.
268: 15983-15993.
Butler, C A. (1996) Lehman Brothers Technical Report "A Paradigm for Success".
Cantrell, Anderson, Cerretti, Price, McKereghan, Tushinski, RJ., Mochizuki, Larsen, Grabstein, L, Gillis, S. and Cosman, D. Proc. Natl. Acad. Sci.
USA 82: 6250-6254.
Cerretti, D.P. et al. (1988) Molecular Immunology 25: 761.
Clang, Rey, Bochner, Heyneker, H. and Gray, G. (1987) Gene: 189-196.
Cotes, P.M. and Bangham, D.R. (196 1) Nature 191: 1065-1067 Cox, McDermott, Merkel, Stroh, Ko, Squires, Gleason, T.M.
and Russell, D. (1994) Endocrinology 135: 1913-1920.
CI Cunningham, B.C. and Wells, L.A. (1989) Science 244:1081-1085.
Cunninghamn, Jhurani, Ng, P. and Wells, J.A. (1989) Science 243: 1330-1336.
Cunningham, Ultsch, de Vos, Mulkerrin, Clauser, K.R. and Wells, J.A. (199 1) Science 254: 821-825.
D'Andrea, Lodish, H.F. and Wong, G.G. (1989) Cell 57: 277-285.
Davis, Aldrich, Stahl, Pan, Taga, Kishimoto, Ip, N.Y. and Yancopoulus, G.D. (1993) Science 260: 1805-1808.
c-i DeChiara, Erlitz, F. and Tamawskik S.J. (1986) Methods Enzymology, 119: 403-15.
INO de la Liosa, Chene, N. and Martal, J. (1985) FEBS Letts. 191: 211-215 Delorme, Lorenzini, Giffin, Martin, Jacobsen, Boone, Elliot, S. (1992) Biochemistry 31: 9871-9876.
de Sauvage, Hass, Spencer, S.D. et al. (1994) Nature 369: 533-538.
de Vos, Ultsch, M. and Kossiakoff, A.A. (1992) Science 25 5: 306-312.
Devos, Plaetinck, Cheroutre, Simons, Degrave, Tavenier, Remaut, E.
and Fiers, W. (1983) Nucleic Acids Research 11: 4307.
Diederichs, Boone, T. and Karplus, A. (1991) Science 154:1779-1792.
Dorssers, Burger, Bat, Deiwel, Van Kessel, Lowenberg, B. and Wagemnaker, G. (1987) 55: 115-124.
Dube, Fisher, Powell, J.S. (1988) J. Biol. Chem. 263: 17526-17521.
Fish, Baneij ee, Levine, B.L. and Stebbing, N. (1986) Antimicrob. Agents Chemnother. 30: 52-56.
Foster, Sprecher, Grant, Kramer, J.M. et al. (1994) Proc. NatI. Acad. Sci USA 91: 13023-13027.
Fukuda, Sasaki Fukuda, M. (1989) Blood 73: 84"9.
Goeddel, Heyneker, ILL., Hozurni, T. et al., (1979) Nature 281: 544-548.
Goeddel, Yelverton, Ullrich, Heynecker, Miozzari, Holmes, W., Seeburg, Dull, Mat, Stebbing, Crea, Maeda, McCandliss, RL, Sloma, Tabor, Gross, Famnilletti, P.C. and Pestka, S. (1980) Nature 287: 411-416.
Goldwasser, E. and Gross, M. (1975) Methods Eazymology 3 7: 109-12 1.
Goodson, R-J. and Katre, N.V. (1990) Biotechnology 8: 343-346.
Goodwin, Lupton, Schmierer, Hjerrild, Jerzy, Clevenger, Gillis, S., Cosman, D. and Namen, A.E. (1989) Proc. Nail. Acad. Sci. USA 86: 302-306.* Gough, N.M. et al., (1988) Proc. Natl. Acad. Sci USA 85: 2623-2627.
Z Gubler, Chua, Schoenbaut, Dwyer, McComas, Motyka, Nabavi, Wolitzky, Quinn, Familletti, P.C. and Gately, M.K. (199 1) Proc. Nat] Acad.
Sci. USA 88: 4143-4147.
sHershfield, Buckley, Greenberg, M.L. et al., (1987) N.Engl. J. Medicine 316: 589-596.
Hill, Osslund, T.D. and Eisenberg, D. (1993) Proc. Natl. Acad. Sci. USA 90:5167-5171.
Hoffian, Andersen, Walker, Kxakover, Patel, Stanmm, M.R. and Osborn, S.0. (1996) Biochemistry 35: 14849-14861.
Hsiung, Mayne, N.G. and Becker, G.W. (1986) Biotechnology 4: 99 1-995.
Hercus, Bagley, Cambareri, Dottore, Woodcock, Vadas, M.A., Shannon, and Lopez, A.F. (1994) Proc. Nail. Acad. Sci. USA 91: 5838-5842.
Hirano, Yasukawa, Harada, Taga, Watana e, Matsuda, Kashiwamura, Nakajima, Koyama, Iwaxnatsu, Tsunasawa, Sakiyamna, Matsui, i s Takahara, Taniguchi, T. and Kishimoto, T. (1986) Nature 324: 73-76.
Horisberger, M.A. and Di Marco, S. (1995) Pbarmac. Ther. 66: 507-534.
Innis, Gelfand, Sninsky, J.J. and White, T.J. (1990) PCR Protocols: A Guide to Methods and Applications. Academic Press, San Diego, CA.
Jacobs, K, Shoemaker, Rudersdorf, R. et al., (1985) Nature 313: 806-8 Karpusas, Nolte, Benton, Meier, Lipscomb, W.N. and Goelz, S. (1997) Proc. Natl. Acad. Sci. USA 94: 11813 -11818.
Katre, N.V. (1990) J. Immunology 144: 209-213.
Kafre, Kaauf, M.J. and Laird, W.J. (1987) Proc. Natl. Acad. Sci. USA 84:1487-1491.
Kawasaki, E.S. et al., (1985) Science 230: 291.
Kawashixna, Obsumi, Mita-Honjo, Shimoda-Takano, Ishikawa, Sakakibara, S., Miyadai, K. and Takiguchi, Y. (199 1) FEBS Letts. 283: 199-202.
Kingsley, D. M. (1994) Genes Dev. 8: 13 3-146.
Komatsu, Nakauchi, Mwa, Isbihara, Eguguchi, M. et al., (199 1) Cancer Research 51, 34 1-348.
3o Kruse, Tony, and Sebald, W. (1992) EMBSO J. 11: 3237-3244.
Lam, Fuller, Miller, Kioss, Manthorpe, Varon, S. and Cordell, B. (1991) Gene 102: 271-276.
CI Kubota, Orita, Hattori, Oh-eda, Ochi, N. and Yamazaki, T. (1990) J. Biochem.
107: 486-492.
Kuga, Komatsu, Yaniasaki, De4kine, Mviyaji, I-i, Nishi, Sato, Yokoo, Asano, Qkabe, Morimoto, M. and Itoh, S. (1989) Bioch.. Biophys. Res. Comm.
159: 103-111.
Kunkel, Roberts, J.D. and Zakour, R.A. (1987) Methods in Enzymology 154: 367-382.
Lange, Valtieri, Santoli, Caracciolo, Mavilio, Gemperlein, Griffin, C., Emanuel,B., Finan, Nowell, P. and Rovera, G. (198 7) Blood 70: 192-199.
Lee, Yokota, Otsuka, Gemmell, Larson, Luh, Arai, and Rennick, D.
(1985) Proc. Natl. Acad. Sci. USA 82: 4360-4364.
Lewis, J.A. (1995) Chapter 9: Antiviral activity of cytokines. pp. 129-14 1.
Li, C.H. (1982) Mol. Cell. Biochem. 46: 31-41.
Lin, (1996) United States Patent 5,547,933.
Lini, Suggs, Lini, et al., Proc. Natl. Acad. Sci. USA (1985) 82: 7580-7584.
Linsley, Kallesta4, Ochs, V. and Neubauer, M. (1990) 10: 1882-1890.
Livnah, Stura, Johnson, D.L. et al., (1996) Science 273: 464-471.
Lu, Clogston, Narhi, Merewether, Pearl, W.R and Boone, T.C. (1992) J. Biol. Chem. 267: 8770-8777.
Lydon, Favre, Bore, Neyret, Benureau, Levine, Seelig, G.F., Nagabhushan, T. L. and Trotta, P.P. (1985) Biochemistry 24: 4131-41.
MacGillivray, Baptista, J1. and Johnson, A. (1996) J. Cliii. Endocrinol. Metab. 8 1: 1806-1809.
M"lk Kallestad, Gunderson, Austin, Neubauer, Ochs, V., Marquardt, Hi., Zarlung, Shoyab, Wei, Linsley, P.S. and Rose, T.M. (1989) Molecular and Cellular Biology 9: 2847-2853.
Martial, Chene, N. and de la Liosa, P. (1985) FEBS. Letts. 180: 295-299.
Martial, Hallewell, ILA., Baxter, J.D. and Goodman, H.M.L (1979) Science 205: 602- 606.
Matthews, Topping, Cass, R.T. and Giebel, L.B. (1996) Proc. Natl. Acad. Sci.
USA 93: 9471-9476.
McKay, D.B. (1992) Science 257: 412.
61 CI McKenzie, Culpepper, Malefyt, R. de Briere, Punnonen, Aversa, G., Sato, Dang, Cooks, Menon, de Vries, Banchereau, and Zurawski, G.
IND (1993) Proc. Nad. Acad. Sci. USA 90: 3735-3739.
Meyers, Paradise, Scudder, Goodman, G. and Konrad, M. (199 1) Guin.
SPharinacol. Ther. 49: 307-313.
Milburn, Hassell, KM., Lambert, Jordan, Proudfoot, Graber, P. and Wells, T.N.C. (1993) Nature 363: 172-176.
Mills, Kostyo, Reagan, Wagner, Moseley, MMH. and Wilhelm, A.E.
IND (1980) Endocrinology 107: 391-399.
Minty, Chalon, Derocq, Dumont, Guilemot, Kaghad, Labit, C., Leplatois, Liauzun, Miloux, M inty, Casellas, Loison. Lupker, Shire, Ferrara, P. and Caput, D. (1993) Nature 362: 248-250.
Moreau, Donaldson, Bennett, Witek-Giannotti, Clark, S.C. and Wong, G.G.
(1988) Nature 336: 690-692.
is Mott, H.R. and Campbell, 1.D. (1995) Current Opinion in Structural Biology 5: 114-121.
Martial, Hailewell, Baxter, J.D. and Goodman, H.M. (1979) Science 205: 602- 606.
Nagata, S. (1994) in Cytokines and Their Receptors, N.A. Nicola ed., Oxford University Press, Oxford, pp. 158-160.
Nagata, Tsuchiya, Asano, Kziro, Yamnazaki, Yamamoto, Hirata, Y., Kubota, Oh-eda, Nomura, H. and Ono, M. (1 986a) Nature 319: 415-418.
Nagata, Tsuchiya, Asano, Yamamoto, Hirata, Kubota, Ob-eda, M., Nomura, H. and Yamazaki, T. (I1986b) EMBO J. 5: 575-58 1.
O'Reilly, DRL, Miller, Luckow, V.A. (1992) Caculovirus Expression Vectors, W.H.
Freeman Co. publishers, New York.
Otsuka, Miyajimna, Brown, Otsu, Abramns, J1., Saeland, Caux, De Waal- Malefyt, De Vries, Meyerson, Yokota, Gernmell, Remnick, Lee, Arai, N., Arai, K.-L and Yokota, T. (1988) J. Immunology 140: 2288-2295.
Owers-Narhi, Arakawa, Aoki, Elmore, Rohde, Boone, Strickland, T.W. (199 1) J. Biol. Chem. 266: 23022-23026.
Ozes, Reiter, Klein, Blatt, L.M. and Taylor, M. W. (1992) J. Interferon Research 12: 55-59.
Paonessa, Gmaziaxi, de Seria, Savino, Ciapponi, Lahm, Ssalvati, A.L., Toniatti, C. and Ciliberto, G. (1995) EMflO J. 14: 1942-1951.
Park, Waldron, Friend, Sassenfeld, Price, Anderson, Cosman, D., Andrews, Bernstein, I.D. and Urdal, D.L. (1989) Blood 74: 56-65.
Paul. Bennett, Calvetti, Kelleher, Wood, O'Hara, Leary, A.C., Sibley, Clark, William, D.A. and Yang, (1990) Proc. Natl. Acad. Sci. USA 87: 7512-7516.
.Pestka, Langer, Zoon, K.C. and Samuel, C.E. (1987) Ann. Rev. Biochem. 56: 727- 777.
riio Pickering, Kronenberg, L.H. and Stewart, 11 (1980) Proc. NatI. Acad. Scio. USA 77: 5938-5942.
Powers, Garrett, March, Frieden, Gronenborn, A.M. and Clore, G.M.
(1992) Science 256:1673-1677.
Radhakrishnan, Walter, Hruka, Reichert, Trotta, Nagabhushan, T. L. and Walter, M.R. (1996) Strucure 4: 1453-1463.
Read, Tomas, Howarth, Martin, Edson, Gillespie, Owens, P.C. and Ballard, F.J. (1992) 2. Endocrinology 133: 421-43 1.
Redfield, Smith Boyd, Lawrence, Edwards, Smith, RXAG., and Dobson, C.M. (199 1) Biochemistry 3 0: 11029-1103 2o Robinson, Grey, L.Ml., Stauton, Vankelecom, Vernalis, Moreau, Stuart, Heath, J.K. and Jones, E.Y. (1994) Cell 77: 1101-1116.
Sasaki, Bothner, Dell, A. and Fukuda, M. (1987)1J. Biol. Chem. 262: 12059-12076.
Sasaki, H. Ochi, Dell, A. and Fukuda, M. (1988) Biochemistry 27: 8616-8626.
Shaw, Veldman, G. and Wooters, J.L. (1992) United States Patent 5,166,322.
Shirafuji, Asano, Matsuda, Watari, Takaku, F. and Nagata, S. (1989) Exp Henatol. 17: 116-119.
Silvennoinen, 0. and IhIe, E.N. (1996) Signalling by the Hematopoietic Cytokine Receptors, R.G. Landes, Company, Austin, TX.
Souza, Boone, Gabrilove, Lai, Zsebo, 1CM., Murdock, Chazin, Bruszewski, Lu, Chen, Barendt, J1., Platzer, Moore, M.A.S., Mertelsmann, R- and Welte, K. (1986) Science 232: 61-65.
Spivak, Hogans, B.B. (1989) Blood 73: 90-99.
CI Takeuchi, Takasaki, Miyazaki, Takashi, Hoshi, Kochibe, N. and Kotaba, A.
Z (1988)1J. Biol. Chem. 263: 3657-3663.
IND Tanaka, Satake-Ishikawa, RK, Ishikawa, Matsuki, S. and Asano, K. (199 1) Cancer Research 51: 3710-3714.
Taniguchi, Masui, Fujita, Takaoka, Kashima, Yoshimoto, R. and Haniuro, 3. (1983) Nature 302: 305-3 Taniguchi, Ohno, Fujii-Kuriyama, Y. and Muraxnatsu., M. (1980) Gene 10: 11 Tarnowski, S.J, Roy, Liptak, Lee, and Ning, R. Y. (1986) Methods Enzymology 119:153-165.
io Tavernier, Tuypens, Verhee, Plaetinck, Devos, Van Der Heyden, Guisez, Y. and Oefner, C. (1995) Proc. Natl. Acad. Sci. USA 92: 5194-5198.
Teh, and Chapman, G.E. (1988) Biochem. Biophys. Res. Comm. 150: 391-398.
Thatcher, D.R. and Panayotatos, N. (1986) Methods Enzymnology 119: 166-177.
Tomnas, Knowles, Owens, Chandler, Francis, Read, L.C. and Ballard, F.J. (1992) Biochem. J. 282: 9 1-97.
Tsuchiya, Asano, Kaziro, Y and Nagata, S. (19 86) Proc. Natl. Acad. Sci. USA 83; 7633-7637.
Tsuda, Kawanishi, Ueda, Masuda, S. and Sasaki, R. (1990) Eur. J. Biochem. 188: 405-411.
Vieira, de Waal-Malefyt, Dang, Johnson, Kastelein, Fiorentino, DRF, de Vries, Roncarolo, Mosmnn, T.R. and Moore, L.W. (199 1) Proc. Natl. Acad.
Sci. USA 88: 1172-1176.
Wang, Lu, S. and Mark, D.F. (1984) Science 224: 1431-1433.
Walter, Cook, Ealick, SM., Nagabhusan, Trotta, P.T. and Bugg, C.E. (1992) J.Mol. Biol. 224:1075-1085.
Wen, Boissel, R, Tracy, Gruninger, Mulcahy, Czelusniak, J., Goodman, M. and Bunn, H.P. (1993) Blood: 1507-1516.
Wen, Boissel, Showers, Ruch, B.C. and Bunn, H.F. (1994) J. Biol. Chem. 269: 22839-22846.
White, B. (1993), Methods in Molecular Biology, volume 15: PCR Protocols: Current Methods and Applications. Humana Press, Totowa, NJ.
Wojchowski, Orkin, Sytkowshi, A.J. (1987) Biochiin. Biophys. Acta 910: 224- 232.
Wolf, Temple, Kobayashi, Young, Dicig, Lowe, Dzialo, Fitz, Ferenz, Hewick, Kelleher, Herrmann, Clark, Azzoni, L., Chan, Trinchieri, G. and Perussia, B. (1991) J. Immunology 146: 3074-3081.
Wong, G.G. et al., (1987) Science 235: 1504.
Wrighton, Farrell, F.X, Chang, R. et al., (1996) Science 273: 458-463.
Yamaguchi, Akai, Kawanisbi, 0. Ueda, Masuda, Sasaki, R (199 1. Biol.
Chem. 266: 20434-20439.
C1 io Yang, Ciarletta, Temple, Chung, Kovacic, Witek-Giannotti, J.S., Leary, Kriz, K, Donahue, ILE, Wong, 0.0. and Clark, S.C. (1986) Cell 47: 3-10.
Yang, Ricciadi, Ciarletta, Calvetti, Kelleher, K. and Clark, S.C. (1989) Blood 74: 1880-1884.
Yokota, Otsuka,, Mosmann, Banchereau, DeFrance, Blanchard, De Vries, Lee F. and Arai, (1986) Proc. Natl. Acad. Sci. USA 83: 5894-5898.
Yokota, Coffmnan, RL, Hagiwara, Rennick, Takebe, Yokota, Gemmell, Shrader, Yang, Meyerson, Luli, Hoy, Pene, Briere, Spits, H., Banchereau, De Vries, Lee, Arai, N. and Arai, K.I. (1987) Proc. Natl. Acad. Sci.
USA 84: 7388-7392.
Zurawski, Imler, and Zurawski, G. (1990) EMvBO J. 9: 3899-3905.
Zurawski, S.M. and Zurawski, 0. (1992) EMBO 1. 11: 3905-3 910.

Claims (22)

1. A cysteine variant of beta interferon (SEQ ID NO:5), wherein at least one cysteine residue is substituted for at least one amino acid located in at least one INO Ci region of the protein selected from the group consisting of: the A-B loop, B-C loop, the C-D loop, the D-E loop, the last three amino acids in helix A, the first three or last three amino acids in helix B, the first three amino acids in helix C, the Sfirst three or last three amino acids in helix D, the first three or last three amino cN acids in helix E, and the amino acids following helix E, or wherein a cysteine Sresidue is added preceding the first amino acid of the mature protein or following N 10 the last amino acid of the protein.
2. The cysteine variant of beta interferon according to claim 1, wherein a cysteine residue is substituted for at least one amino acid selected from the group consisting of: Q23, N25, G26, R27, E29, Y30, K33, D34, R35, N37, D39, E42, E43, K45, Q46, L47, Q48, Q49, Q51, K52, E53, F70, R71, Q72, D73, S74, S76, T77, G78, N80, E81, T82, K108, E109, D110, F111, T112, R113, G114, K115, L116, A135, K136, E137, K138, S139, L160, T161, G162, Y163, L164, R165 and N166.
3. The cysteine variant of beta interferon according to claim 2, wherein a cysteine residue is substituted for at least one amino acid located in the region of beta interferon following helix E.
4. The cysteine variant according to claim 3, wherein a cysteine residue is substituted for at least one amino acid selected from the group consisting of Y163, L164, R165 and N166. The cysteine variant of beta interferon according to claim 1, wherein a cysteine residue is substituted for at least one amino acid located in the A-B loop of beta interferon.
6. The cysteine variant according to claim 5, wherein a cysteine residue is substituted for at least one amino acid selected from the group consisting of: Q23, 00 O N25, G26, R27, E29, Y30, K33, D34, R35, N37, D39, E42, E43, K45, Q46, L47, cN Q48, and Q49.
7. The cysteine variant according to claim 1, wherein a cysteine residue is ID cN substituted for at least one amino acid located in the B-C loop of beta interferon. S 5 8. The cysteine variant according to claim 7, wherein a cysteine residue is Ssubstituted for at least one amino acid selected from the group consisting of: Q72, D73, S74, S75, S76, T77 and G78.
9. The cysteine variant of beta interferon according to claim 1, wherein a cysteine residue is substituted for at least one amino acid located in the C-D loop of beta interferon. The cysteine variant according to claim 9, wherein a cysteine residue is substituted for at least one amino acid selected from the group consisting of: K108, E109, D110, Fl11, T112, R113, G114, K115, and L116.
11. The cysteine variant of beta interferon according to claim 1, wherein a cysteine residue is substituted for at least one amino acid located in the D-E loop of beta interferon.
12. The cysteine variant according to claim 11, wherein a cysteine residue is substituted for at least one amino acid selected from the group consisting of E137 and K138.
13. The cysteine variant of beta interferon according to claim 1, wherein a cysteine residue is substituted for at least one amino acid located in at least one region of beta interferon selected from the group consisting of the first three amino acids in helix B or the last three amino acids in helix B.
14. The cysteine variant according to claim 13, wherein a cysteine residue is substituted for at least one amino acid selected from the group consisting of: Q51, K52, E53, F70 and R71. 00 The cysteine variant of beta interferon according to claim 1, wherein a C cysteine residue is substituted for at least one amino acid selected from the group Sconsisting of the first three amino acids in helix C of beta interferon. C 16. The cysteine variant according to claim 15, wherein a cysteine residue is substituted for at least one amino acid selected from the group consisting of: E81 and T82.
17. The cysteine variant of beta interferon according to claim 1, wherein a Icysteine residue is substituted for at least one amino acid located in at least one Sregion of beta interferon selected from the group consisting of the first three amino acids in helix D or the last three amino acids in helix D.
18. The cysteine variant according to claim 17, wherein a cysteine residue is substituted for at least one amino acid selected from the group consisting of Al and K136.
19. The cysteine variant of beta interferon according to claim 1, wherein a cysteine residue is substituted for at least one amino acid located in at least one region of beta interferon selected from the group consisting of the first three amino acids in helix E or the last three amino acids in helix E. The cysteine variant according to claim 19, wherein a cysteine residue is substituted for at least one amino acid selected from the group consisting of S139, L160, T161 and G162.
21. The cysteine variant of beta interferon according to claim 1, wherein a cysteine residue is added preceding the first amino acid of the mature protein of beta interferon.
22. The cysteine variant of beta interferon according to claim 1, wherein a cysteine residue is added following the last amino acid of beta interferon-2. 00 O 23. The cysteine variant of beta interferon according to any one of claims 1 to N 22, wherein a non-cysteine amino acid is substituted for C17.
24. The cysteine variant of beta interferon according to any one of claims 1 to cI 22, wherein a serine residue is substituted for C17.
25. The cysteine variant of beta interferon according to any one of claims 1 to 22, wherein an alanine residue is substituted for C17.
26. The cysteine variant according to any one of claims 1 to 25, wherein the substituted or added cysteine residue is modified with a cysteine-reactive moiety.
27. The cysteine variant according to any one of claims 1 to 25, wherein the substituted or added cysteine residue is modified with a polyethylene glycol.
28. A method to treat a condition that is treatable with beta interferon, comprising administering the cysteine variant according to any one of claims 1 to 27 to a patient with said condition.
29. Use of a cysteine variant according to any one of claims 1 to 27 in a medicament for the treatment of a condition treatable with beta interferon. BOLDER BIOTECHNOLOGY, INC. WATERMARK PATENT TRADE MARK ATTORNEYS P16775AU02 c1 SEQUENCE LISTING <110> Cox III, George N Bolder Biotechnology, Inc. <120> Derivatives of Growth Hormone and Related Proteins <130> BBO011 <140> unknown <141> 1998-07-14 <150> 60/052,516 <151> 1997-07-14 <160> 41 <170> Patentln Ver. <210> 1 <211> 191 <212> PRT <213> Homo sapiens <400> 1 Phe Pro Thr Ile Pro Leu Ser Arg Leu Phe ASp Asn Ala Met Leu Arg 1 5 10 Ala His Arg Leu His Gin Leu Ala Phe Asp Thr Tyr Gin Giu Pbe Glu 25 Glu Ala Tyr Ile Pro Lye GIu GIn Lys Tyr Ser Phe Leu Gin Asn Pro 40 Gin Thr Ser Leu Cys Phe Ser Glu Ser Ile Pro Thr Pro Ser As Arg 55 Glu Giu Thr Gin Gin Lye Ser Asn Leu Glu Leu Leu Arg Ile Ser Leu 70 75 Leu Leu Ile Gin Ser Trp Leu Glu Pro Val Gln Phe Leu Arg Ser Val 90 Phe Ala Asn Ser Leu Val Tyr Giy Ala Ser Asp Ser Asn Val Tyr Asp 100 105 110 Leu Leu Lye Asp Leu Giu Glu Gly Ile Gin Thr Leu Met Gly Arg Leu 115 120 125 Glu Asp Gly Ser Pro Arg Thr Gly Gin lie Phe Lys Gin Thr Tyr Ser 130 135 140 Lye Phe Asp Thr Asn Ser His Asn Asp Asp Ala Leu Leu Lye Asn Tyr 145 150 155 160 Gly Leu Leu Tyr Cyz Phe Arg Lye Asp Met Asp Lys Val Glu Thr Phe 165 170 175 Leu Arg lie Val Gin Cys Arg Ser Vai Giu Gly Ser CyB Gly Phe 180 185 190 IND IND <210> 2 <211.> 166 <212> PRT <213> Homno sapiens <400> 2 Ala Pro Pro Arg Lou Ile Cys Asp 1. 5 Leu Giu Ala Lys Glu Ala Glu Asn Cys Ser Leu Asn Giu Asn Ile Thr 35 40 Tyr Ala Trp Lys Arg Met Giu Vai 55 Gin Gly Lou Ala Leu Leu Ser Giu 70 Lou Val Asa Ser Ser Gin Pro Trp Lys Ala Val Ser Gly Leu Arg Ser 100 Gly Ala Gin Lys Giu Ala Ile Sex 115 120 Pro Leu Arg Thr Ile Thr Ala Asp 1.30 135 Tyr Ser Asa Phe Leu Arg Gly Lys 145 ISO Cys Arg Thr Giy Asp Arg 165 Scr Ile 25 Val Gly Ala Glu Leu 105 Pro Tbr Leu Arg 10 Thr Pro Gin Val Pro 90 Thr Pro Phe Lys Vai Thr Asp Gin Leu 75 Leu Thr Asp Arg Lau 155 Leu Giy Thr Ala Arg Gin Leu Ala Lys 140 Glu Arg Tyr Leu is Cys Ala Giu His Lys Val Asn Phe Val Giu Val Trp, Gly Gin Ala Leu Leu His Val Asp Leu Arg Ala Leu .110 Ala Ser Ala Ala 2S Leu Phe Arg Vai Thr Gly Giu Ala 160 <210> 3 <211> 165 <212> PRT <213> Homo sapiens <400> 3 Cys Asp Lou Pro Gin Tbx His Ser Lou Gly 1 5 10 Lou Leu Ala Gin Met Arg Arg Ile Ser Lou 25 Arg His Asp Phe Gly Phe Pro Gin Giu Giu 40 Lys Ala Giu Thr Ile Pro Val Leu His Glu 55 Asn Leu Phe Ser Thr Lys Asp Ser Ser Ala 70 Ser Arg Arg Thr Lou Met Phe Ser Cys Leu Lys Asp Phe Gly Asn Gin Phe Gin Met Ile Gin Gin Ile Phe Ala Trp Asp Giu Thr Leu 7S s0 IND IND Leu Ala Glu Tyr Ala 145 Leu Phe Ile 100 Ile Glu Met Lys Thr Oiu Lou Tyr Gin Gin Leu Asn Asp Leu Glu 90 Gly Val Gly Val Thr Giu Thr Pro Leu Met Lys 105 110 Ala Val Arg Lys Tyr Phe Gin Azg Ile Thr Leu 120 125 Lye Tyr Ser Pro Cys Ala Trp Giu Vai Val Arg 135 140 Ser Phe Ser Leu Ser Thr Asm LeU Gin Giu Ser 150 155 160 c210> 4 <211> 166 <212> PRT <213> HoMO sapiens <400> 4 Cys Asp Leu Pro Giu Thr His Ser Leu Asp Ann Arg Arg Thr Leu Met 1 5 10 Leu Leu Ala Oln Met ser Arg Ile Set Pro Ser Ser Cys Lau Met Asp 25 Arg His Asp Phe Gly Pie Pro Gin Giu Giu Pie Asp Gly Ann Gln Phe 40 Gin Lye Ala Pro Ala Ile Ser Val Lau His Glu Lou lie Gin Gin Ile SO 55 Phe Ann Leu Phe Tbr Thr Lye Asp Set Ser Ala Ala Trp Asp Glu Asp 70 75 Leu Leu Asp Lye Phe Cya Thr Giu Lou Tyr Gin Gin Leu Asn Asp Leu 90 Glu Ala Cys Val Met Gin Giu Giu Arg Val Gly Giu Thr Pro Lou Met 100 105 110 Asn Ala Asp Ser lie Leu Ala Val Lye Lye Tyr Phe Arg Arg lie Thr 115 120 125 Leu Tyr Leu Thr Giu Lys Lys Tyr Ser Pro Cys Ala Trp Glu Val Val 130 135 140 Arg Ala Giu Ile Met Arg Ser Lou Set Lou Set Thr Ann Lou Gin Glu 145 150 155 160 Arg Lou Axg Arg Lye Glu 165 c210> IND IND <211> 166 <212> PRT <213> Homo sapiens <400> 5 Met Ser Tyr Asn Leu Leu Gly Phe Leu Gin 1 5 10 Cys Gin Lys Leu Leu Trp Gin Leu Aan Gly 20 25 Lys Asp Arg Met Asn Phe Asp le Pro Glu 40 Gin Phe Gin Lys Giu Asp Ala Ala Leu Thr 55 Asn Ile Phe Ala Ile Phe Arg Gin Asp Ser 70 Giu Thr Ile Val Giu Asn Lou Leu Ala Asn 90 His Lou Lys Thr Val Leu Giu Glu Lys Leu 100 105 Arg Gly Lys Leu Met 6cr Sor Leu His Leu 115 120 Ile Leu His Tyr Leu Lys Ala Lys Giu Tyr 130 135 lie Val Arg Vai Glu Ile Leu Arg Asn Phe 145 150 Thr Gly Tyr Leu Arg Asn 165 Arg Ser Ser Arg Leu Glu Glu Ile Lys Ile Tyr Glu Ser Ser Thr 75 Val Tyr His Giu Lys Glu Lys Arg Tyr 125 Ser His Cys 140 Tyr Phe Ile 155 Asn Pbe Gln 15 Tyr Cys Leu Gin Leu Gln Met Leu Gin Gly Trp Asn Gin Ile Asn Asp Phe Thr 110 Tyr Gly Arg Ala Trp Thr Asn Arg Leu 160 <210> 6 <211> 174 c212> PRT <213> Homo sapiens <400> 6 Thr Pro Leu Gly Pro Ala Ser Ser Leu Pro Gin Ser Phe Leu Leu LyS 1 S 10 is Cys Lou Giu Gin Vai Arl Lys Ile Gin Gly Asp Gly Ala Ala Lou Gin 25 Glu Lys Leu Cys Ala Thr Tyr Lys Leu Cys His Pro Giu Giu Leu Val 40 Leu Leu Gly His Ser Lou Giy Ile Pro Trp Ala Pro Leu Ser Ser Cys 55 Pro Ser Gin Ala Lou Gin Lou Ala Giy Cys Lou Ser Gin Lou His Ser 70 75 IND IND Ieu Leu Asp Thr 105 Gin Met 120 Ala Met Leu Val Leu Arg Gin Ala Leu Glu Gly Leu Gin Leu Asp Val 110 Glu Glu Leu Gly Met 125 Pro Ala Phe Ala Ser 140 Ala Ser His Leo Oln 155 His Leu Ala Gin Pro 170 Ile Ser Ala Asp Ala Pro Ala Phe Ser Phe 160 c210> 7 <211> 332 <212> PRT <213> Homo sapiens <400> 7 Ser Pro Ala Pro Pro Ala Cys Asp 1 Arg Asp His Pro Gly Glu Gly Ala Leu Gly Val Arg Pro Pro Leu Ser 130 His Pro Lys Thr Thr Leu 100 Gly Gin Leo Axg Val Leu 10 Arg Leu 3cr Gn 25 Ser Lys Leu Leo Cys Pro Glu Val Val Leu His Ser Thr Thr Leo Cys Leu Arg His Pro Val Gin Met Leu Leu 70 Leu er Gly Ala Thr Thr Leu Leu 135 Leu Leu Pro Ala Val Asp Phe Ser Leo 40 Glu Glu Glu Gly Ser Leo Leu Gin 105 Ala His 120 Arg Gly Lys Met 75 Gly Leu Asp Val Ala 155 Gin Asp Ala Arg Leo Ser Gly Thr 110 Asa Ala 125 Phe Leo Pro Thr Leo Gin (3n Leu Phe Le Ala 160 Val Gly Gly Ser Thr Leo Cys Val Arg Val Pro Ser Arg Thr Ser Leo Val Leo Thr Leo Asn Glu Leu Pro Asn 165 170 175 Ala Arg Thr 190 Arg Thr Ser Gly 180 Leu Leo Glu Thr Asa phe 185 Thr Ala Ser IND IND Thr Gly Pro Gly 210 Tyr Leu 225 Pro Gly Thz Ser Pro Ser Pro Pro 290 Asp Pro 305 Ser Tyr Ser Gly 195 Leu LeU Asf Arg Pro Ser Asp Thr 260 Pro Thr 275 Thr Leu Ser Ala Thr His Leu Lys Trp Gin Gin Gly Phe 200 Gln Thr Ser Arg Ser LeU Asp 215 220 His Glu Leu Leu Asn Gly Thr 230 235 Arg Thr Leu Gly Ala Pro Asp 250 Ser Leu Pro Pro Asn Leu Gin 265 Pro Pro Thr Giy Giy Tyr Thx 280 Thr Pro Val Val Gin Leu His 295 300 Thr Pro Thr Pro Thr Ser Pro 310 315 Gin Asn Leu Ser Gin Giu Gly 330 Arg Ala Lys le 205 Gin Ile Pro Gly Arg Gly Leu Phe 240 Ile Ser Scr Gly 255 Pro Gly Tyr Ser 270 Leu Phe Pro Leu 285 Pro Leu Leu Pro Leu Leu Asn Thr 320 <210> 8 <211> 127 <212> PRT <213> Homo sapiens <400> 8 Ala Pro Ala Arg Ser 1 5 Ann Ala Ile Gin Glu Ala Ala Glu Met Asn Leu Gin Giu Pro Thr Gly Leu Arg Gly Ser Ala Ser His Tyr Lys Ala Thr Gin Ile Ile 100 Phe Leu Leu Val Ile 115 Pro Arg Val 40 Gin Lys ys Glu Asp 120 Thr Gin Pro Trp 10 Leu Ann Leu Ser Val Ile Ser Glu Arg Leu Glu Leu Lys Gly Pro Leu 75 Pro Thr Pro Glu 90 Phe Lys Giu A=n Trp Giu Pro Val 125 Glu His Val Arg Asp Thr Met Phe Asp Tyr Lys Gin Thx Met Met Thr Ser Cys Leu Lys Asp 110 Gin Glu <2105 9 IND <211> 133 <212> PRT <213> Howo sapiens <400> 9 Ala Pro Thr Ser Ser 1 5 Len Leu Leo Asp Leo Asn Pro Lys Lau Thr Lys Ala Thr Glu Leu 50 Pro Leu Giu Glu Val Arg Pro Arg ASp Leu Lys Gly Ser Giu Thr 100 Thr Ile Val Glu Phe 115 Ile Scr Thr Leu Thr 130 Ser Thr Lye Lye Thr 10 Gln met Ile Leu Asn 25 Arg Met Leu Thr Phe 40 Lys His Len Gin Cys 55 Leo Asn Leo Ala Gin 70 Ile ger Ann Ile Asa 90 Thr Phe Met Cys Glu 105 Leu Asn Arg Trp Ile 120 Gin Leu Gin Leu Gbu His Gly Ile Asn Asn Tyr Lys Lys Phe Tyr Met Pro LYS Leu Glu Glu Gin Leu Lys Ser Lys Asn Phe His Lou 75 B0 Val Ile Vai Len Gin Leo Tyr Ala Asp Giu Thr Ala 11.0 Thr Phe eye Gin 5cr Ile 125 <210> <211> 152 <212> PRT <213> Hoamo sapiens <400> 2.0 Met Ser Arg Lou Pro Val 1 5 Gly Len Gin Ala Pro Met Val Ann eye Ser Asia Met Pro Pro Len Pro Len LeU so Asp Ilie Leu Met Glu An 70 Asn Arg Ala Val Lys Ser Leu Lye Ann LeU Lou Pro 100 Arg His Pro Ile His Ile Leu Leu Gin 10 Thr Thr Pro 25 Gi Ile Ile Asn Asn Leu Arg Arg Pro 75 Asn Ala Ser 90 Pro Leu Ala 105 Gly Asp Trp Lau Leo Thr An An Ala Thr Asa Leu Val Arg Pro is Lye Tbr Ser TrP His Lou Lys Gin Gly Gin Asp Gin Leu Giu Ala Phe Ile Gin Ser Ile Ala Ala Pro Thr 110 Glu Phe Arg Arg c1 115 120 125 Lys Leu Thr Phe Tyr Leu Lys Thr Leu Giu Asn Ala Gin Ala Gin Gin 130 135 140 Thr Thr Leu Ser Leu Ala Ile Phe 145 150 <210> 11 0\ <211> 129 <212> PRT <213> Homo sapiens <400> 11 His Lys Cys ASp Ile Thr Leu Gin Glu Ile Ile Lye Thr Leu Asn Ser 1 5 10 Leu Thr Giu Gin Lye Thr Leu Cys Thr Glu Leu Thr Val Thr Asp Ile 25 Phe Ala Ala Ser Lys Asn Thr Thr Giu Lye Glu Thr Phe Cys Arg Ala 40 Ala Thr Val Leu Arg Gin Phe Tyr Ser His His Glu Lys Asp Thr Arg 55 Cys Leu Gly Ala Thr Ala Gin Gin Phe His Arg'His Lye Gin Leu Ile 70 75 Arg Phe Leu Lys Arg Leu Asp Arg Asn Leu Trp Gly Leu Ala Giy Len 90 Asn Ser Cys Pro Val Lys Glu Ala Asn Gin Ser Thr Leu Glu Asn Phe 100 105 110 Leu Giu Arg Leu Lye Thr Ile Met Arg Glu Lye Tyr Ser Lye Cys Ser 115 120 125 Ser <210> 12 <211> 134 4212> PRT <213> Homo sapiens <400> 12 Met Arg Met Leu Leu His Leu Ser Leu Leu Ala Leu Gly Ala Ala Tyr 1 5 10 Val Tyr Ala Ile Pro Thr Giu Ile Pro Thr Ser Ala Leu Val Lys Glu 25 Thr Leu Ala Leu Leu Ser Thr His Arg Thr Leu Leu Ile Aia Asn Glu 40 Thr Leu Arg lie Pro Val Pro Val His Lye Asn His Gin Leu Cys Thx s0 55 IND IND Giu Glu Ile Phe Gin Giy Ile Gly Thr Leu Glu 3cr Gin Thr Val Gin 70 75 Gly Gly Thr Vai Giu Arg Leu Phe Lys Asn Leu Ser Leu Ile Lys Lys 90 Tyr Ile Asp Gly Gin Lys Lys Lys Cys Gly Giu Glu Arg Arg Arg Val 100 105 110 Aen Gin Phe Leu Asp Tyr Leu Gln Giu Phe Leu Gly Val Met Aen Thr 115 120 125 Giu TrP Ile Ile Glu Ser 130 <210> 13 <211> 212 <212>. PIZT <213> Homo sapiens <400> 13 Met Asn Ser Phe Se 1 Gly Leu Leu Leu Va. Gly Glb ASP Scr Ly Ser Scr Giu Arg 11 so Ser Ala Leu Arg L2 8cr Lys Giu Ala LE Giu Lys Asp Gly C: 100 Val Lys Ile Ile 115 Leu Gin Asn Arg P 130 Met 3cr Thr Lys V 145 Leu Asp Ala Ile T I Thr Lys Leu Gin 180 r 5 ye r he al Thr Ser Ala Phe Gly Pro Val Ala PheSe5r Leu 10 Leu Pro Ala Ala Phe Pro Ala Pro Val Pro Pro 25 Asp VJal Ala Ala pro His Arg Gin Pro Leil Thr 40 Asp Lys Gin Ile Arg Tyr Ile Leu Asp Gly Ile 55 G2.u Thr Cys Asn Lye Ser Asn Met Cys Giu Ser 70 75 Ala Glu Ann Asn Leo Ann Leu Pro Lys Met Ala 90 Phe Gin Ser Gly Phe Asn Giu Giu Thr Cys Leo 105 110 Gly Leu Leu Glu Phe Giu Val Tyr Leu Giu Tyr 120 125 Giu Ser Ser Giu Giu Gin Ala Arg Ala Val Gin 135 140 Lei Ile Gin Phe Lcu Gin Lys Lye Ala Lye Asn 150 155 160 Thr Pro Asp Pro Thr Thr Ann Ala Ser Leo Leu 170 175 Gin Ann Gin Trp, Leu Gin Asp Met Tbr Thr His 185 190 C IND Leu Ile Leu Arg Ser Phe Lys Glu Phe Leu Gin Ser Ser Leu Arg Ala 195 200 205 Leu Arg Gin Met 210 <210> 14 <211> 177 <212> PRT <213> Homo sapiens <400> 14 Met Phe His Val Ser Phe Arg Tyr 1 S Leu Val Leu Leu Pro Vai Ala Ser Asp Gly Lys Gin Tyr Glu Scr Val 40 Leu Asp Ser Met Lye Giu Ile Gly 55 Asn Phe Phe Lye Arg His Ile Cys 70 Leu Phe Arg Ala Ala Arg Lys Leu Thr Giy Asp Phe Asp Leu His Leu 100 Ile Leu Leu Asn Cys Thr Gly Oln 115 120 Leu Giy Giu Ala Gin Pro Thr Lys 130 135 Lye Glu Gln Lye Lys Leu Asn Asp 145 150 Gin Giu Ile Lye Thr Cys Trp Asn 165 His Ile Ser 25 Leu Ser Asp Arg Leu 105 Val Ser Leu Lye Phe Gly Leu 10 Asp Cys Asp Met Val Ser Asn Cya Leu Ala Aen Lye 75 Gln Phe Leu 90 Lys Val Ser Lys Giy Arg Leu Glu Glu 140 Cys Phe Leu 155 Ile Leu Met 170 Pro Pro Leu Ile Ile Giu Gly Lye Ile Asp Gin Leu Asn Asn Glu Phe Giu Gly Met Phe Lye Met Aen Ser Glu Gly Thr Thr 110 Lye Pro Ala Ala 12S Asn Lye Ser Leu Lys Arg Leu Leu 160 Gly Tbr Lys Glu 175 <210> <211> 144 <212> PRT <213> Homo sapiens <400> Met Leu Leu Ala Met Val Leu Tbr Ser Ala Leu Leu Leu Cys Ser Val 1 5 10 Ala Gly Gin Gly Cys Pro Thr Leu Ala Gly Ile Leu Asp Ile Ann Phe 25 IND IND Leu Ile Agn Ala Ann Val Thr Arg Pro 65 Met Gin Thr Glu Val Leu Cys Asn Gin 115 Leu Giu Ile 130 Glu Asp 40 Leu Cys 55 Glu Arg Leu Ile Lye Cys Gly Asn 120 Glu Lye 135 Ala Ser Lye Cys His Cya Gly Ile Pro Ser Asp Asn Ser Gin Met Thr Ann Thr Ser Arg Val Lye Lys Ser 90 Tyr Phe Ser Cys Giu Gin 110 Leu Thr Phe Leu Lys Ser 125 Arg Gly Met Arg Gly Lys 140 <210> 16 <211> 178 <212> PRT c213, Homo sapiens <400> 16 Met His Ser Ser Ala 1 5 Arg Ala Ser Pro Gly Phe Pro Gly Asn Leu Ser Arg Val Lye Thr Leu Leu Lye Glu Ser Gin Ala Leu Ser Qlu Gln Ala Glu Ann Gin 100 Gly Giu Ann Leu Lys 115 Phe Leu Pro Cys Glu 130 Ala Phe Asn Lys Leu 145 jeu Gla Pro Phe Leu 70 Met Amp Thr Asn Gln 150 Leu Cys Cys Gly Thr Gln 25 Ann Met Leu 40 Phe Gin Met 55 Leu Glu Asp Ile Gln Phe Pro Asp Ile 105 Leu Arg Leu 120 Lye Ser Lys 135 Glu Lye Gly Leu Ser Arg Lys Phe Tyr 90 Lye Arg Ala Ile Val Lou Leu Thr Glu An Ser Cys Asp Leu Arg Asp. Asp Gin Lou Asp Lye Gly Tyr Leu 75 Lau Giu Glu Val Ala His Val Asn 110 Leu Arg Arg Cys 125 Val Giu Gin Val 140 Tyr Lye Ala met 155 Gly Vai rhr His Ala Phe Asn Leu Gly Cys so Met Pro Ser Leu His Arg Lye Asn ser Glu 160 Phe Asp Ile Phe Ile Asn Tyr Ile Glu Ala Tyr Met Thr Met Lys Ile 165 170 175 Arg Asn <210> 17 <211> 199 <212> PRT <213> Homo sapiens S<400> 17 Met Asn Cys Val Cys Arg Leu Val Leu Val Val Leu Ser Leu Trp Pro 1 5 10 (71 Asp Thr Ala Val Ala Pro Gly Pro Pro Pro Gly Pro Pro Arg Val Ser 25 Pro Asp Pro Arg Ala Glu Lou Asp Ser Thr Val Leu Leu Thr Arg Ser 40 Leu Leu Ala Asp Thr Arg Gln Leu Ala Ala Gln Leu Arg Asp Lys Phe 55 Pro Ala Asp Gly Asp His Asn Leu Asp Ser Leu Pro Thr Leu Ala Met 70 75 Ser Ala Gly Ala Leu Gly Ala Leu Gin Lou Pro Gly Val Leu Thr Arg 90 Leu Arg Ala Asp Leu Leu Ser Tyr Leu Arg His Val Gln Trp Leu Arg 100 105 110 Arg Ala Gly Gly Ser Ser Leu Lys Thr Leu Glu Pro Glu Leu Gly Thr 115 120 125 Leu Gin Ala Arg Leu Asp Arg Leu Leu Arg Arg Leu Gln Leu Leu Met 130 135 140 Ser Arg Leu Ala Leu Pro Gln Pro Pro Pro Asp Pro Pro Ala Pro Pro 145 150 155 160 Leu Ala Pro Pro Ser Ser Ala Trp Gly Gly Ile Arg Ala Ala His Ala 165 170 175 Ile Leu Gly Gly Leu His Lou Thr Leu Asp Trp Ala Val Arg Gly Leu 180 185 190 Leu Leu Leu Lys Thr Arg Leu 195 <210> 18 <211> 219 <212> PRT <213> Homo sapiens <400> 18 Met Cys Pro Ala Arg Ser Len Len Leu Val Ala Thr Leu Val Leu Leu 1 5 10 IND Asp His Len Ser Leu Ala Arg Asn Leu Pro Val Ala Thr Pro Asp Pro 25 Gly Met Phe Pro Cys Leu His His Ser Gin Asn Leu Leu Arg Ala Val 40 'Ser Asn Met Leu Gln Lys Ala Arg Gin Thr Leu Giu Phe Tyr Pro Cys 55 Thr Ser Gu Glu Ile Asp His Gin Asp Ile Thr Lys Asp Lys Thr Ser \O 65 70 75 Thr Val Glu Ala Cys Leu Pro Leu Glu Le Thr Lys Asn Glu Ser Cys 90 Leu Asn Ser Arg Giu Thr Ser Phe Ile Thr Asn Gly Ser Cys Leu Ala 100 105 110 Ser Arg Lys Thr Ser Phe Met Met Ala Leu Cys Leu Ser Ser Ile Tyr 115 120 125 Glu Asp Leu Lys Met Tyr Gin Val Giu Phe Lys Thr Met Asn Ala Lys 130 135 140 Len Len Met Asp Pro Lys Arg Gn Ile Phe Leu Asp Gin Asn Met Len 145 150 155 160 Ala Val Ile Asp Glu Len Met Gin Ala Len Asn Phe Asn Ser Gin Thr 165 170 175 Val Pro Gin Lys Ser Ser Leu Giu Giu Pro Asp Phe Tyr Lys Thr Lys 180 185 190 Ile Lys Leu Cys Ile Len Leu His Ala Phe Arg Ile Arg Ala Val Thr 195 200 205 le Asp Arg Val Thr Ser Tyr Leu Ann Ala Ser 210 215 <210> 19 <211> 132 <212> PRT <213> Homo sapiens <400> 19 Met Ala Len Len Leu Thr Thr Val Ile Ala Len Thr Cys Len Gly Gly 1 5 10 Phe Ala Ser Pro Gly Pro Val Pro Pro Ser Thr Ala Len Arg Glu Len 25 Ile Giu Glu Leu Val Asn Ile Tbr Gin Asn Gin Lys Ala Pro Len Cys 40 Asn Giy Ser Met Val Trp Ser Ile Asn Leu Tbr Ala Gly Met Tyr Cys 55 IND IND Ala Ala Lou Glu Ser Leu Ile Asn Val Ser Gly Cys Ser Ala Ile Glu 70 75 s0 Lys Thr Gin Arg Met Leu Ser Gly Phe Cys pro His Lys Val Ser Ala BS 90 Gly Gin Phe Ser Ser Lou His Val Arg Asp Thr Lys Ile Glu Val Ala 100 105 110 Gin Phe Val Lys Asp Leu Lou Lou His Lou Lys Lys Leu Phe Arg Giu 115 120 125 Gly Arg Phe AMn 130 <210> <211> 114 <212> PRT <213> HOMO sapiens <400> 20 AMn Trp Val Msn Val 1 5 Gin Ser Met His Ile Pro Ser Cys Lys Vai Val Ile Ser Lou Glu Asn Leu Ilie Ile Leu Thr Giu Ser Gly Cys Lys Glu Phe Leu Gin 100 Thr Ser Ser Asp Lou Lys Lys Ala Thr Leu Tyr Thr 25 Ala Met Lys Cys Phe 40 Gly Asp Ala Ser Ile 55 Asn Asn Ser Leu Ser Giu Cys Giu Giu Leu 90 Phe Val His Ile Val 105 Ile Glu Asp Lou Giu Ser Asp Vai Lau Lou Giu Lou His Asp Thr Val Ser Asn Gly Ass Giu Giu Lys Msn Gin Met Phe Ile 110 <210> 21 <211> 252 <212> PRT <213> Homo sapiens <400> 21 met Giy Val Lou Leu Thr Gin Axg Thx Leu Leu Ser Leu Val Leu Ala 1 5 10 Leu Leu Phe Pro Ser Met Ala 8cr Met Ala Ala Ile Gly Ser Cys Ser 25 14 Lys Glu Tyr Arg Val Leu Leu Gly 40 Gin Lou Gin Lye Gin Thr Asp Leu IND IND Met Leu Phe Gin Leu Asr Gly 145 Asp Gin Asp Pro Thr Glu Ile 130 Leu Thr Asp Val Ser Leu Gin 115 Glu Arg Ala Thr Pro Glu Asn 100 Arg Asp Aen Glu Leu Glu Arg Gly Ala 120 Leu Cys Ala Asp His Gly Cys 105 Gin Gin Met Gly Pro ys Leu Val Aap Met Ala Arg 170 Tyr Arg 75 Gly Leu Leu Ala Gin iss oly Ile Giu Arg His Glu Arg 140 Leu Ala Arg Arg Arg Arg Arg 125 Pro Leu Ser Ile Pro Gly Leu 110 Ser Asn Asp Gin Gin Gly Phe Ala Gly Ile Aen Pro 175 Gly Gly Ala Leu Asp Leu Leu Sex 160 Pro Cys Thr Pro Thr Pro Ala Ser Asp Ala Phe Gin Arg Lye Leu Glu 180 185 190 His Gly Trp Gly Lau Arg Leu Met 245 Arg Phe 200 Pro Asn Val Arg Gly Gin His Ber Val 205 Ser Arg Arg 220 Thr Arg Pro 235 Pro Arg Gly Arg His Ser Ser Arg Val Pro Lye '240 4210> 22 <211> 200 <212> PRT c213> Homo sapiens <400> 22 Met Ala Phe Tbr Glu His Ser Pro Lou Thr Pro His Arg Arg Asp Leu 1 5 10 Cys Ser Arg Ser Ile Trp Lou Ala Arg Lys Ile Arg Ser Asp Lou Thr 25 Ala Leu Thr Glu Sex Tyr Val Lys His Gin Gly Leu Asn Lys Asn Ile 40 Asn Lou Asp Sex Ala Asp Gly Met Pro Val Ala Sex Thr Asp Gin Trp 55 Ser Arg RigB Leu Leu Asfl 145 Val Arg Tyr Leu Thr Giu Ala Giu Phe His Val Leu Leu Thr Pro Tbhr Glu Gly 100 Gin Val Ala Ala Phe 115 Glu Tyr Lys Ile Pro 135 Gly Asp Gly Gly Leu 150 Gin Glu Leu Ser Gin 165 Ile Ser Ser His Gin 180 Ala Ann Asn Lys Lys 195 <210> 23 <211> 181 <212> PRT <213> HOMO Sapiens (400> 23 Ser Pro Leu Pro Ile 1 5 Pro Cys His Ann An Leu Ann Giy Ser Ala Gly Giu Pro Phe Pro Thr Asp Phe Pro Pro Val Giu Leu Tyr Arg Ile Thr Arg Asp Gin 100 Ser Lys Leu Asn Ala 115 Val Leu Cys Arg Leu Val Ann Ala Ann Gin Ile Leix Phe Ile 40 Leu Asp Lys Ala Asn Gly Val Tyr Leu Leu Ann Pro 105 Asp Ile Leu 120 Lys Tyr His Cys Ala Ser Gin Tyr Tyr Cys, Gly Giu Lys Thr Ser Ala Leu Giy Leu 125 Giy His His Gin Gin Vai Leu An His An Val C1130 135 140 Thr Tyr Gly Pro Pro Asp Thr ser Gly Lys Asp Val Phe Gin Lys Lys 145 150 155 160 C1Lys Lau Gly Cys Gin Leu Lou Gly Lys Tyr Lys Gin Ile Ile Ala Val 165 170 175 Lou Ala Gin Ala Phe 180 <210> 24 INC <212> DNA <213> Artificial Sequence (NI 220>* <223> Description of Artificial Sequence:PCR Primer <400> 24 gggggtcgac catatgttcc caaccattcc cttatccag 39 <210> <211> 33 <212> DNA <213> Artificial Sequence <220> c223> Description of Artificial Sequence:PCR Primer <400> gggggatcct cactagaagc cacagctgcc ctc 33 <210> 26 <211> 39 c212> DNA <213> Artificial Sequence <220> <223> Description of Artificial Sequence:PCR Primer <400> 26 ccccggatcc gccaccatgg atctctggca gctgctgtt 39 <210> 27 c211> <212> DNA <213> Artificial Sequence <220> <223> Description of Artificial SequencesPCR Primer <400> 27 ccccgtcgac tctagagcta. ttaaatacgt agctcttggg '<210> 28 '<211> 32 '<212> DNA <213> Artificial Sequence CK1 <220> <223> Description of Artificial Sequence:PCR Primer <400> 29 cgcggatccg attagaatcc acagctcccc tc 32 <210> 29 c211> 66 I><212>o DNA <213> Artificial Sequence <220> c223> Description of Artificial Sequence:PCR Primer INO <400> 29 ccccctctag acatatgaag aagaacatcg cattcctgct, ggcatctatg ttcgttttct ctatcg 66 <210> <211> <212> DNA <213> Artificial Sequence <220> <223> Description of Artificial Sequence:PCR Primer <400> 30 gcatctatgt tcgttttctc tatcgctacc aacgcttacg cattcccaac cattccctta tccag <210> 31 <211> 62 <212> DNA <213> Artificial Sequence <220> <223>. Description of Artificial Sequence:PCR Primer <400> 31 gcagtggcac tggctggttt cgctaccgta gegcaggcct tcccaaccat tcccttatcc ag 62 <210> 32 <211> 59 <212> DNA 4213>. Artificial Sequence c220> <223> Description of Artificial Sequence:PCR Primer <400> 32 ccccgtcgac acatatgaag aagacagcta tcgcgattgc agtggcactg gctggtttc 59 <210> 33 <211> 36 <212> DNA <213> Artificial Sequence <220> <223> Description of Artificial Sequence:PCR Primer (N2~ <400> 33 __ctgcttgaag atctgcccac accgggggct gccatc 3 <210> 34 <211> 24 <212> DNA <213> Artificial Sequence <220> <223> Description of Artificial Sequence:PCR Primer <400> 34 gtagcgcagg ccttcccaac catt 24 <210> INO <211> 39 <212> DNA <213> Artificial Sequence <220> <223> Description of Artificial Sequence:PCR Primer <400> ctgcttgaag atctgcccag tccgggggca gccatcttc 39 <210> 36 <211> 51 <212> DNA <213> Artificial Sequence c220> <223> Description of Artificial Sequence:PCR Primer <400> 36 gggcagatct tcaagcagac ctacagcaag ttcgactgca actcacacaa c 51 <210> 37 <211> 34 <212> DNA <213> Artificial Sequence <220> <223> Description of Artificial Sequence:PCR Primer <400> 37 cgcggtaccc gggatccgat tagaatccac agct 34 <210> 38 <211> 36 D<212> DNA <213> Artificial Sequence c220> <223> Description of Artificial Sequence:PCR Primer <400> 38 gggcagatct tcaagcagac ctactgcaag ttcgac 36 <210> 39 <211> 42 <212> DNA
AU2006203197A 1997-07-14 2006-07-26 Derivatives of growth hormone and related proteins Ceased AU2006203197B2 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
AU2006203197A AU2006203197B2 (en) 1997-07-14 2006-07-26 Derivatives of growth hormone and related proteins
AU2008203051A AU2008203051B2 (en) 1997-07-14 2008-07-10 Derivatives of growth hormone and related proteins

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US60/052516 1997-07-14
AU2002306305A AU2002306305C1 (en) 1997-07-14 2002-11-28 Derivatives of growth hormone and related proteins
AU2006203197A AU2006203197B2 (en) 1997-07-14 2006-07-26 Derivatives of growth hormone and related proteins

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
AU2002306305A Division AU2002306305C1 (en) 1997-07-14 2002-11-28 Derivatives of growth hormone and related proteins

Related Child Applications (1)

Application Number Title Priority Date Filing Date
AU2008203051A Division AU2008203051B2 (en) 1997-07-14 2008-07-10 Derivatives of growth hormone and related proteins

Publications (2)

Publication Number Publication Date
AU2006203197A1 AU2006203197A1 (en) 2006-08-17
AU2006203197B2 true AU2006203197B2 (en) 2008-04-10

Family

ID=36888684

Family Applications (1)

Application Number Title Priority Date Filing Date
AU2006203197A Ceased AU2006203197B2 (en) 1997-07-14 2006-07-26 Derivatives of growth hormone and related proteins

Country Status (1)

Country Link
AU (1) AU2006203197B2 (en)

Also Published As

Publication number Publication date
AU2006203197A1 (en) 2006-08-17

Similar Documents

Publication Publication Date Title
AU751898B2 (en) Derivatives of growth hormone and related proteins
US8148500B2 (en) Method of preparing cysteine mutants of human recombinant GM-CSF
AU2006203197B2 (en) Derivatives of growth hormone and related proteins
AU2008203051B2 (en) Derivatives of growth hormone and related proteins
AU2002306305C1 (en) Derivatives of growth hormone and related proteins

Legal Events

Date Code Title Description
FGA Letters patent sealed or granted (standard patent)
MK14 Patent ceased section 143(a) (annual fees not paid) or expired