CA2472085A1 - N-terminally chemically modified protein compositions and methods - Google Patents

N-terminally chemically modified protein compositions and methods Download PDF

Info

Publication number
CA2472085A1
CA2472085A1 CA002472085A CA2472085A CA2472085A1 CA 2472085 A1 CA2472085 A1 CA 2472085A1 CA 002472085 A CA002472085 A CA 002472085A CA 2472085 A CA2472085 A CA 2472085A CA 2472085 A1 CA2472085 A1 CA 2472085A1
Authority
CA
Canada
Prior art keywords
protein
csf
monopegylated
pegylated
polyethylene glycol
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
CA002472085A
Other languages
French (fr)
Inventor
Olaf B. Kinstler
Nancy E. Gabriel
Christine E. Farrar
Randolph B. Deprince
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.)
Amgen Inc
Original Assignee
Amgen Inc.
Olaf B. Kinstler
Nancy E. Gabriel
Christine E. Farrar
Randolph B. Deprince
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=23250903&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=CA2472085(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Amgen Inc., Olaf B. Kinstler, Nancy E. Gabriel, Christine E. Farrar, Randolph B. Deprince filed Critical Amgen Inc.
Publication of CA2472085A1 publication Critical patent/CA2472085A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/56Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule
    • A61K47/59Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyureas or polyurethanes
    • A61K47/60Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyureas or polyurethanes the organic macromolecular compound being a polyoxyalkylene oligomer, polymer or dendrimer, e.g. PEG, PPG, PEO or polyglycerol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/06Antihyperlipidemics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/06Antianaemics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/53Colony-stimulating factor [CSF]
    • C07K14/535Granulocyte CSF; Granulocyte-macrophage CSF
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/555Interferons [IFN]
    • C07K14/56IFN-alpha
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides

Abstract

Provided herein are preparations and compositions relating to the attachment of water soluble polymers to proteins. The water soluble polymer can be polytheylene glycol conjugated to the protein via an amino group on the protein, wherein the protein is monopegylated and the polyethylene glycol is conjugated predominantly at the N-terminus of the protein.

Description

-l N-TERMINALLY CHEMICALLY MODIFIED PROTEIN
COMPOSITIONS AND METHODS
Fi e1 c~ o f the Inv,~,ntion The present invention broadly relates to the field of protein modification, and, more specifically, the attachment of water soluble polymers to proteins or analogs thereof (the term "protein" as used herein is synonymous with "polypeptide" or "peptide" unless otherwise indicated). The present invention also relates to novel methods for N-terminally modifying proteins or analogs thereof, and resultant compositions.
In another aspect, the present invention relates to novel N-terminally chemically modified G-CSF
compositions and related methods of preparation. The present invention also relates to chemically modified consensus interferon.
B~aclcaround Proteins for therapeutic use are currently available in suitable forms in adequate quantities largely as a result of the advances in recombinant DNA
technologies. The availability of 'recombinant proteins has engendered advances in protein formulation and ,chemical modification. One goal of such modification is protein protection. Chemical attachment may effectively block a proteolytic enzyme from physical contact with .
the protein backbone itself, and thus prevent degradation. Additional advantages include, under certain circumstances, increasing the stability and circulation time of the therapeutic protein and decreasing immunogenicity. A review article describing protein modification and fusion proteins is Francis, Focus on Growth Factors ~: 4-10 (May 1992) (published by Mediscript, Mountview Court, Friern Barnet Lane, London N20, OLD, UK) .
Polyethylene glycol ("PEG") is one such chemical moiety which has been used in the preparation of therapeutic protein products (the verb "pegylate"
meaning to attach at least one PEG molecule). For example Adagen, a pegylated formulation of adenosine deaminase is approved for treating severe combined immunodeficiency disease; pegylated superoxide dismutase has been in clinical trials for treating head injury;
pegylated alpha interferon has been tested in phase I
clinical trials for treating hepatitis; pegylated glucocerebrosidase and pegylated hemoglobin are reported to have been in preclinical testing. The attachment of polyethylene glycol has been shown to protect against proteolysis, Sada, et al., J. Fermentation Bioengineering u,: 137-139 (1991), and methods for attachment of certain polyethylene glycol moieties are available. ~, U.S. Patent No. 4,179,337, Davis et al., "Non-Immunogenic Polypeptides," issued December 18, 1979; and U.S. Patent No. 4,002,531, Royer, "Modifying enzymes with Polyethylene Glycol and Product Produced Thereby," issued January 11, 1977. For a review, see Abuchowski et al., yn Enzymes as Drugs. (J. S.
Holcerberg and J. Roberts, eds. pp. 367-383 (1981)).
Other water soluble polymers have been used, such as copolymers of ethylene glycol/propylene glycol, carboxymethylcellulose, dextrin, polyvinyl alcohol, polyvinyl pyrrolidone, poly-1, 3-dioxolane, poly-1,3,6-trioxane, ethylene/maleic anhydride copolymer, polyaminoacids (either homopolymers or random copolymers).
For polyethylene glycol, a variety of means have been used to attach the polyethylene glycol molecules to the protein. Generally, polyethylene glycol molecules are connected to the protein via a reactive group found on the protein. Amino groups, such a CA 02472085 2004-07-08 as those on lysine residues or at the N-terminus, are convenient for such attachment. For example, Royer (U. S. Pat. No. 4,002,531, above) states that reductive alkylation was used for attachment of polyethylene glycol molecules to an enzyme. EP 0 539 167, published April 28, 1993, Wright, "Peg Imidates and Protein Derivates Thereof" states that peptides and organic compounds with free amino groups) are modified with an immediate derivative of PEG or related water-soluble organic polymers. U.S. Patent No. 4,904,584, Shaw, issued February 27, 1990, relates to the modification of the number of lysine residues in proteins for the attachment of polyethylene glycol molecules via reactive amine groups.
One specific therapeutic protein which has been chemically modified is granulocyte colony stimulating factor, "G-CSF." G-CSF induces the rapid proliferation and release of neutrophilic granulocytes to the blood stream, and thereby provides therapeutic effect in fighting infection.
European patent publication EP 0 401 384, published December 12, 1990, entitled, "Chemically Modified Granulocyte Colony Stimulating Factor,"
describes materials and methods for preparing G-CSF to which polyethylene glycol molecules are attached.
Modified G-CSF and analogs thereof are also reported in EP 0 473 268, published March 9,' 1992, entitled "Continuous Release Pharmaceutical Compositions Comprising a Polypeptide Covalently Conjugated To A
Water Soluble Polymer," stating the use of various G-CSF
and derivatives covalently conjugated to a water soluble particle polymer, such as polyethylene glycol.
A modified polypeptide having human granulocyte colony stimulating factor activity is reported in EP 0 335 423 published October 4, 1989.
Another example is pegylated IL-6, EP 0 442 724, entitled, "Modified hIL-6, " (see co-pending U.S.S.N. 07/632,070) which discloses polyethylene glycol molecules added to IL-6.
EP 0 154 316, published September 11, 1985 reports reacting a lymphokine with an aldehyde of polyethylene glycol.
Many methods of attaching a polymer to a protein involve using a moiety to act as a linking group. Such moieties may, however, be antigenic. A
tresyl chloride method involving no linking group is available, but this method may be difficult to use to produce therapeutic products as the use of tresyl chloride may produce toxic by-products. ~ Francis et al., In: Stability of protein pharmaceuticals: in vivo pathways of degradation and strategies for protein stabilization (Eds. Ahern., T. and Manning, M.C.) Plenum, New York, 1991) Also, Delgado et al., "Coupling of PEG to Protein By Activation With Tresyl Chloride, Applications In Immunoaffinity Cell Preparation", Fisher et al., eds., Separations Using Aqueous Phase Systems, Applications In Cell Biology and Biotechnology, Plenum Press, N.Y.N.Y.,1989 pp. 211-213.
Chamow et al., Bioconjugate Chem. ~: 133-140 (1994) report the modification of CD4 immunoadhesin with monomethoxlypoly(ethylene glycol) aldehyde via reductive alkylation. The authors report that 50% of the CD4-Ig was MePEG-modified under conditions allowing the control over the extent of pegylation. ~. at page 137. The authors also report that the ~ vitro binding capability of the modified CD4-Ig (to the protein gp 120) decreased at a rate correlated to the extent of MePEGylation.
Ibid. ~ also, Rose et al., Bioconjugate Chemistry ~,:
154-159 (1991) which reports the selective attachment of the linker group carbohydrazide to the C-terminal carboxyl group of a protein substrate (insulin).
None of the methods in the general state of the art, or the art relating to particular proteins, allow for selective attachment of a water soluble polymer to the N-terminus of a protein such as G-CSF, however. Rather, the currently existing methods provide for non-selective attachment at any reactive group, whether located within the protein, such as a lysine side group, or at the N-terminus. This results in a heterogenous population. For example, for pegylated G-CSF molecules, some molecules have a different number of polyethylene glycol moieties than others. As an illustration, protein molecules with five lysine residues reacted in the above methods may result in a heterogenous mixture, some having six polyethylene glycol moieties, some five, some four, some three, some two, some one and some zero. And, among the molecules with several, the polyethylene glycol moieties may not be attached at the same location on different molecules.
This is disadvantageous when developing a therapeutic.pegylated protein product. In such development, predictability of biological activity is crucial. For example, it has been shown that in the case of nonselective conjugation of superoxide dismutase with polyethylene glycol, several fractions of the modified enzyme were completely inactive (P.McGoff et al. Chem. Pharm. Bull. x:3079-3091 (1988)). One cannot have such predictability if the therapeutic protein differs in composition from lot to lot. Some of the polyethylene glycol moieties may not be bound as stably in some locations as others, and this may result in such moieties becoming dissociated with the protein. Of course, if such moieties are randomly attached and therefore become randomly dissociated, the pharmacokinetics of the therapeutic protein cannot be precisely predictable. From a consumer's point of view, the circulation time may vary from lot to lot; and thus dosing may be inaccurate. From a producer's point of view, garnering regulatory approval for sale of the therapeutic protein may have added complexities.
Additionally, none of the above methods provide for selective N-terminal chemical modification without a linking moiety (between the protein and the polymer).
If a linking moiety is used, there may be disadvantages due to possible antigenicity.
Thus, there exists a need for methods allowing for selectively N-terminally chemically modified proteins and analogs thereof, including G-CSF and consensus interferon (two chemically modified proteins exemplified below), The present invention addresses this need in a number of aspects.
~mma y of the Invention The present invention relates to substantially homogenous preparations of N-terminally chemically modified proteins, and methods therefor. Unexpectedly, chemical modification at the N-terminus of G-CSF
demonstrated advantages in stability which are not seen in other G-CSF species containing one chemical modification at another location on the molecule. Also unexpectedly, in the present process for making N-terminally chemically modified G-CSF, it was found that using reductive alkylation, one could provide conditions for selectively modifying the N-terminus, and this method is broadly applicable to other proteins (or analogs thereof), as well as G-CSF. Also surprisingly, using reductive alkylation,~the end product -- protein with an amine linkage to the Water soluble polymer --was found to be far more stable than identical -polymer/protein conjugate having an amide linkage. One other protein so modified (as described in a working example below) is consensus interferon. Thus, as described below in more detail, the present invention has a number of aspects relating to chemically modifying proteins (or,analogs thereof) as well as specific modifications of specific proteins.
In one aspect, the present invention relates to a substantially homogenous preparation of N-terminally chemically modified G-CSF (or analog thereof) and related methods. One Working example below demonstrates that N-terminally monopegylated G-CSF more stable than other types of monopegylated G-CSF.
Additionally, since the N-terminus of the G-CSF molecule is more available during reaction with polyethylene glycol, a higher proportion of the N-termini are pegylated, and therefore, this species provides processing advantages.
The present invention also relates to a type of reductive alkylation which selectively activates a-amino group of the N-terminal residue of a protein or analog thereof, thereby providing for selective attachment of a water soluble polymer moiety at the N-terminus. This provides for a substantially homogenous preparation of polymer/protein conjugate molecules as well as (if polyethylene glycol is used) a preparation of pegylated protein molecules having the polyethylene glycol moiety directly coupled to the protein moiety. This method is described below for G-CSF and for consensus interferon, and these provide for additional aspects of the present invention.

l ) - 8 _ FIGURE lA is a reproduction of the chromatogram of the peaks from ion exchange chromatography of pegylated G-CSF.
FIGURE 1B is an SDS-PAGE of various species of mono-pegylated G-CSF.
FIGURE 2 is an SEC-HPLC profile of (Line A) recombinant human methionyl G-CSF standard; (Line B) SCM-PEG-GCSF reaction mix; SLine C) N-terminally pegylated~G-CSF; (Line D) lysine 35 monopegylated G-CSF;
(Line E) lysine 41 monopegylated G-CSF.
FIGURES 3A, 3B, and 3C are HPLC endoproteinase SV8 peptide mapping tracings of (3A) N-terminally pegylated G-CSF; (3B) lysine 35 monopegylated G-CSF;
(3C) lysine 41 monopegylated G-CSF.
FIGURE 4 is a bar graph illustrating a comparison of y~ vitro bioactivity of monopegylated G-CSF species compared to an unpegylated standard.
FIGURES 5A and 5B are graphs illustrating results of ~1 viyo bioactivity assays of monopegylated G-CSF derivatives, with 5A illustrating the average hamster white blood cell count after a single subcutaneous injection of N-terminally pegylated G-CSF, lysine 35 monopegylated G-CSF, or lysine 41 monopegylated G-CSF, and 5B illustrating the net average white blood cell count area under the curve after a single subcutaneous injection of the various ' monopegylated G-CSF derivatives~listed above.
FIGURES 6A, 6B, and 6C are SEC-HPLC profiles for stability studies of N-terminally pegylated G-CSF or lysine 35 monopegylated G-CSF. FIGURES 6A and 6B are the profiles for stability studies conducted at pH 6.0 at 4°C for (.6A)-N-terminally monopegylated G-CSF or (6B) lysine 35 monopegylated G-CSF. FIGURE 6C shows the profiles for extended stability studies at pH 6.0 and _ 9 _ 4°C for lysine 35 monopegylated G-CSF. Time ("T") indicates days.
FIGURE 7 illustrates size exclusion HPLC
analysis of the reaction mixture in the process of reductive alkylation of rh-G-CSF with methoxypolyethylene glycol aldehyde (MW 6 kDa).
FIGURE 8 illustrates size exclusion HPLC
analysis of the reaction mixture using N-hydroxysuccinimidyl ester of 1~EG, also at MW=6kDa.
. FIGURE 9 illustrates the total white blood cell response after a single subcutaneous dose to mono-N
terminal I~EG-GCSF conjugates prepared by reductive alkylation of rh-G-CSF with I~EG aldehydes of different molecular weights (6 kDa,l2kDa and 20 kDa).
petailed Description The present invention relates to substantially homogenous preparations of N-terminally chemically modified proteins, and methods therefor.
In one aspect, the present invention relates to N-terminally chemically modified G-CSF compositions and methods therefor.
The present methods (for both N-terminally modified G-CSF as well as the present reductive alkylation methods) provide for a substantially homogenous mixture of monopolymer/protein conjugate.
"Substantially homogenous" as used herein means that the only polymer/protein conjugate~molecules observed are those having one polymer moiety. The preparation may contain unreacted (i.e., lacking polymer moiety) protein. As ascertained by peptide mapping and N-terminal sequencing, one example below provides for a preparation which is at least 90% monopolymer/protein conjugate, and at most 10% unreacted protein.
Preferably, the N-terminally monopegylated material is at least 95% of the preparation (as in the working example below) and most preferably, the N-terminally monopegylated material is 99% of the preparation or more. The monopolymer/protein conjugate has biological activity. The present "substantially homogenous"
N-terminally pegylated G-CSF preparations provided herein are those which are homogenous enough to display the advantages of a homogenous preparation, e.g., ease in clinical application in predictability of lot to lot pharmacokinetics.
One may choose to prepare a mixture of polymer/protein conjugate molecules, and. the advantage provided herein is that one may select the proportion of monopolymer/protein conjugate to include in the mixture.
Thus, if desired, one may prepare a mixture of various protein with various numbers of polymer moieties attached (i.e., di-, tri-, tetra-, etc.) and combine with the monopolymer/protein conjugate material prepared using the present methods, and have a mixture with a predetermined proportion of monopolymer/protein conjugate.
Provided below is a working example using G-CSF, which, as described above, is a therapeutic protein used to treat hematopoietic disorders. In general, G-CSF useful in the practice of this invention may be a form isolated from mammalian organisms or, alternatively, a product of chemical synthetic procedures or of prokaryotic or eukaryotic host expression of exogenous DNA sequences obtained by genomic or cDNA cloning or by DNA synthesis. Suitable prokaryotic hosts include various bacteria (e. g., . coli); suitable eukaryotic hosts include yeast (e. g., , c,~revis'iae) and mammalian cells (e. g., Chinese hamster ovary cells, monkey cells). Depending upon the host employed, the G-CSF expression product may be glycosylated with mammalian or other eukaryotic carbohydrates, or it may be non-glycosylated. The G-CSF
expression product may also include an initial methionine amino acid residue (at position -1). The present invention contemplates the use of any and all such forms of G-CSF, although recombinant G-CSF, especially ~ ~ derived, is preferred, for, among other things, greatest commercial practicality.
Certain G-CSF analogs have been reported to be biologically functional, and these may also be chemically modified, by, for example, the addition of one or more polyethylene glycol molecules. G-CSF
analogs are reported in U.S. Patent No. 4,810,643.
Examples of other G-CSF analogs which have been reported to have biological activity are those set forth in AU-A-76380/91, EP 0 459 630, EP 0 272 703, EP 0 473 268 and EP 0 335 423, although no representation is made with regard to the activity of each analog reportedly disclosed. ~ also AU-A-10948/92, PCT US94/00913 and EP
0 243 153.
Generally, the G-CSFs and analogs thereof useful in the present invention may be ascertained by practicing the chemical modification procedures as provided herein to selectively chemically modify the N-terminal a-amino group, and testing the resultant product for the desired biological characteristic, such as the biological activity assays provided herein. Of~
course, if one so desires when.treating non-human mammals, one may use recombinant non-human G-CSF's, such as recombinant murine, bovine, canine, etc. ~ PCT WO
9105798 and PCT WO 8910932, for example.
Thus, another aspect of the present invention includes N-terminally chemically modified G-CSF analog compositions. As described above, G-CSF analogs may include those having amino acid additions, deletions ) and/or substitutions (as compared to the G-CSF amino acid sequence set forth in Example 1, below). Those G-CSF analogs which are predicted to function when N-terminally pegylated to selectively stimulate the production of neutrophils are those with an N-terminus which is not necessary for binding to a G-CSF receptor.
~ Hill et al., PNAS-USA ~Q: 5167-5171 (1993); ~ also PCT US94/00913.
The polymer molecules used may be selected from among water soluble polymers. (For the reductive alkylation procedure described herein, the polymers should have a single reactive aldehyde.) The polymer selected should be water soluble so that the protein to which it is attached does not precipitate in an aqueous environment, such as a physiological environment. For reductive alkylation, the polymer selected should have a single reactive aldehyde so that the degree of polymerization may be controlled as provided for in the present methods. The polymer may be branched or unbranched. Preferably, for therapeutic use of the end-product preparation, the polymer will be pharmaceutically acceptable. One skilled in the art will be able to select the desired polymer based on such considerations as whether the polymer/protein conjugate will be used therapeutically, and if so, the desired dosage, circulation time, resistance to proteolysis, and other considerations: For G-CSF, these may be ascertained using the assays provided herein, and one skilled in the art should select the appropriate assays for other therapeutic proteins. The water soluble polymer may be selected from the group consisting of, for example, those listed above (in the Background section), and dextran or poly(n-vinyl pyrrolidone)polyethylene glycol, propropylene glycol homopolymers, prolypropylene oxide/ethylene oxide co-polymers, polyoxyethylated polyols and polyvinyl alcohol.
Subject to considerations for optimization as discussed below, the polymer may be of any molecular weight, and may be branched or unbranched. For polyethylene glycol, the preferred molecular weight is between about 2kDa and about 100kDa (the term "about"
indicating that in preparations of polyethylene glycol, some molecules will weigh more, some less, than the stated molecular weight). Examples 1 and 2 below involve the use of PEG 6000, which was selected for ease in purification and for providing an adequate model system. Other sizes may be used, depending on the desired therapeutic profile (e.g., the duration of sustained release desired, the effects, if any on biological activity, the ease in handling, the degree or lack of antigenicity and other known effects of the polyethylene glycol to a therapeutic protein or analog).
One specific aspect of the present invention is N-terminally monopegylated G-CSF comprised of a polyethylene glycol moiety and a G-CSF moiety. For the present compositions, one may select from a variety of polyethylene glycol molecules (by molecular weight, branching, etc.), the proportion of polyethylene glycol molecules to G-CSF protein molecules in the reaction mix, the type of pegylation reaction to be performed, the method of obtaining the selected N-terminally pegylated G-CSF, and the type of G-CSF to be used.
Further, the present compositions and methods include formulation of pharmaceutical compositions, methods of treatment and manufacture of medicaments.
The proportion of polyethylene glycol molecules to protein molecules will vary, as will their concentrations in the reaction mixture. In general, the optimum ratio (in terms of_efficiency of reaction in that there is no excess unreacted protein or polymer) will be determined by the molecular weight of the polyethylene glycol selected. In addition, as one example of the present methods involves non-specific pegylation and later purification of N-terminally monopegylated species, the ratio may depend on the number of available reactive groups (typically ~ or 3 amino groups) available. One working example herein involved a fairly low reaction ratio of protein: PEG
molecules~to obtain monopegylated material generally (1.5 PEG molecules per protein molecules).
For obtaining N-terminally pegylated G-CSF, the method for pegylation may also be selected from among various methods, as discussed above, or the present reductive alkylation as described in Example 2, below. A method involving no linking group between the polyethylene glycol moiety and the protein moiety is described in Francis et al., In: Stability of protein pharmaceuticals: in vivo~pathways of degradation and strategies for protein stabilization (Eds. Ahern., T.
and Manning,-M.C.) Plenum, New York, 1991) Also, Delgado et al., "Coupling of PEG to Protein By Activation With Tresyl Chloride, Applications In Immunoaffinity Cell Preparation", ,~: Fisher et al., eds., Separations Using Aqueous Phase Systems, Applications In Cell Biology and Biotechnology, Plenum Press, N.Y.N.Y.,1989 pp. 211-213, involves the use of tresyl chloride, which results~in no linkage group between the polyethylene.glycol moiety and the protein moiety. This method may be difficult to use to produce therapeutic products as the use of tresyl chloride may produce toxic by-products. One of the present working examples involves the use of N-hydroxy succinimidyl esters of carboxymethyl methoxy polyethylene glycol. As will be discussed in more detail below, another working example involves the use of the present reductive alkylation methods.
The method of obtaining the N-terminally pegylated G-CSF preparation (i.e., separating this moiety from other monopegylated moieties if necessary) may be by purification of the N-terminally pegylated material from a population of pegylated G-CSF molecules.
For example, presented below is an example where pegylated G-CSF is first separated by ion exchange chromatography to obtain material having a charge characteristic of monopegylated material (other multi-pegylated material having the same apparent charge may be present), and then the monopegylated materials are separated using size exclusion chromatography. In this way, N-terminally monopegylated G-CSF was separated from other monopegylated species, as well as other multi-pegylated species. Other methods are reported.
For example, PCT WO 90/04606, published May 3, 1990, reports a process for fractionating a mixture of PEG-protein adducts comprising partitioning the PEG/protein adducts in a PEG-containing aqueous biphasic system.
In a different aspect, the present invention provides a method for selectively obtaining an N-terminally chemically modified protein (or analog).
Provided below is a method of protein modification by reductive alkylation which exploits differential reactivity of different types of primary amino groups (lysine versus the N-terminal)~available for derivatization in a particular protein. Under the appropriate reaction conditions, substantially selective derivatization of the protein at the N-terminus with a carbonyl group containing polymer is achieved. The reaction is performed at pH which allows one to take advantage of the pKa differences between the e-amino groups of the lysine residues and that of the a-amino group of the N-terminal residue of the protein. By such selective derivatization attachment of a water soluble polymer to a protein is controlled: the conjugation with the polymer takes place predominantly at the N-terminus of the protein and no significant modification of other reactive groups, such as the lysine side chain amino groups, occurs.
Importantly, and surprisingly, the present invention provides for a method of making a substantially homogenous preparation of monopolymer/protein conjugate molecules, in the absence of further extensive purification as is required using other chemical modification chemistries. Additionally, the product having an amine linkage is unexpectedly more stable than a product produced with an amide linkage, and this is demonstrated in the aggregation studies below. More specifically, if polyethylene glycol is used, the present invention also provides for N-terminally pegylated protein lacking possibly antigenic linkage groups, and having the polyethylene glycol moiety directly coupled to the protein moiety without toxic by-products.
The reaction may be diagrammed as follows (indicating sodium cyanohydroboride as an illustrative reducing agent):

PROTEIN ~-- NH2 O
I I
H-C-P EG
NaCNBH3 PROTEIN ~-NH-CH2-PEG

__ Thus, one aspect of the present invention is a method for preparing a polymer/protein conjugate comprised of (a) reacting a protein moiety having more than one amino group with a water soluble polymer moiety under reducing alkylation conditions, at a pH suitable to selectively activate the a -amino group at the amino terminus of said protein moiety so that said water soluble polymer selectively attaches to said oc-amino group: and (b) obtaining the reaction product. One may optionally, and preferably for a therapeutic product, separate the reaction products from unreacted moieties.
Another aspect of the present invention is that such reductive alkylation will provide for selective attachment of the polymer to any protein having an a -amino group at the amino terminus, and provide for a substantially homogenous preparation of monopolymer/ protein conjugate. The term "monopolymer/
protein conjugate" is used here to mean a composition comprised of a single polymer moiety attached to a protein moiety (also encompassed are those conjugates using protein analogs as described herein). The monopolymer/protein conjugate will have a polymer moiety located at the N-terminus, but not on amino side groups, such as those for lysine. The preparation will preferably be greater than 80% monopolymer/ protein conjugate, and more preferably greater than 95%
monopolyme~r protein conjugate. ' For a substantially homogenous population of monopolymer/protein conjugate molecules, the reaction conditions are those which permit the selective attachment of the water soluble polymer moiety to the N-terminus of the desired protein. Such reaction conditions generally provide for pKa differences between the lysine amino groups and the oc-amino group at the N-terminus (the pK being the pH at which 50% of the i amino groups are p.rotonated and 50~ are not). In general, for different proteins, different pH's may be used for optimally modifying the a-amino groups of the N-terminus.
The pH also affects the ratio of polymer to protein to be used. In general, if the pH is lower than the pK, a larger excess of polymer to protein will be desired (i.e., the less reactive the N-terminal a-amino group, the more polymer needed to achieve optimal conditions). If the pH is higher than the pK, the polymer: protein ratio need not be as large (i.e., more reactive groups are available, so fewer polymer molecules are needed).
Another important consideration is the molecular weight of the polymer. In general, the higher the molecular weight of the polymer, the fewer number of polymer molecules which may be attached to the protein.
Similarly, branching of the polymer should be taken into account when optimizing these parameters. Generally, the higher the molecular weight (or the more branches) the higher the polymer: protein ratio.
For the present reductive alkylation, the reducing agent should be stable in aqueous solution arid preferably be able to reduce only the Schiff base formed in the initial process of reductive alkylation.
Preferred reducing agents may be selected from the group consisting of sodium borohydride, sodium cyanoborohydride, dimethylamine borate, timethylamine borate and pyridine borate. Sodium cyanoborohydride was used in the working examples below.
The water soluble polymer may be of the type described above, and should have a single reactive aldehyde for coupling to the protein. For polyethylene glycol, use of PEG 6000 for coupling to G-CSF and PEG
12000 for consensus interferon are described below.- It S

is noted, that for G-CSF, PEG 12000, 20000 and 25000 have also been used successfully in the present methods.
Polyethylene glycol propionaldenhyde (see, e.g., U.S.
Patent No. 5,252,714) is advantageous for its stability in water.
As indicated above, the present methods are broadly applicable to any protein or analog thereof having an N-terminal a-amino group. For example, proteins which are the product of an exogenous DNA
sequence 'expressed in bacteria may have, as a result of bacterially expression, an N-terminal methionyl residue with an a-amino group. As indicated above, peptides are included, as are peptidomimetics and other modified proteins. Protein analogs, such as the G-CSF analogs Z5 described above, and the non-naturally occurring consensus interferon are also suitable for the present methods.
Thus, for the present N-terminally chemically modified G-CSF, any of the G-CSF's or analogs as described. herein may be used (e.g.,.those described supra). The working examples below use recombinant G-CSF produced in bacteria, having 174 amino acids and an extra N-terminal methionyl residue. As described herein, the chemical modification may be performed with any of the water soluble polymers described herein, and the present working examples describe the use of polyethylene glycol.
Consensus interferon~is another protein used in the present working examples. Demonstrated below is the preparation. of chemically modified consensus interferon using the pxesent reductive alkylation methods for N-terminal monopegylation. Thus, other aspects of the present invention relate to these preparations. As employed herein, consensus human leukocyte interferon, referred to here as "consensus . - 21 -interferon," or "IFN-con", means a nonnaturally-occurring polypeptide, which predominantly includes those amino acid residues that are common to all naturally-occurring human leukocyte interferon subtype sequences arid which include, at one or more of those positions where there is no amino acid common to all subtypes, an amino acid which predominantly occurs at .
that position and in no event includes any amino acid residue which is not extant in that position in at least one naturally-occurring subtype. IFN-con encompasses the amino acid sequences designated IFN-conl, IFN-con2 and IFN-con3 which are disclosed in commonly owned U.S.
Patents 4,695,623 and 4,897,471.
(U. S. Patent Nos.
4,897,471 and 4,695,623 use the denomination "a" which is not used herein.) DNA sequences encoding IFN-con may be synthesized as described in the above-mentioned patents or other standard methods. IFN-con polypeptides are preferably the products of expression of manufactured DNA sequences, transformed or transfected into bacterial hosts, especially ~. coli. That is, IFN-con is recombinant IFN-con. IFN-con is preferably produced in ~ coli may be purified by procedures known to those skilled in the art and generally described in Klein et al.;J. Chromatog. ~: 205-215 (1988) for IFN-conl. Purified IFN-con may comprise a mixture of isoforms, e.g., purified IFN-conl comprises a mixture of methionyl IFN-conl, des-methionyl, IFN-conl and des-methionyl IFN-conl with a blocked N-terminus (Klein et al., Arc. Biochem. Biophys. ~: 531-537 (1990)).
Alternatively, IFN-con may comprise a specific, isolated isoform. Isoforms of IFN-con are separated from each other by techniques such as isoelectric focusing which are known to those skilled in the art.

Thus, another aspect of the present invention is a chemically modified consensus interferon wherein said consensus interferon moiety is selected from the group consisting of IFN-con y IFN-con2, and IFN-con3.
The chemical modification is using a water soluble polymer as described herein, such as PEG, and the present reductive alkylation methods may be used for selective N-terminal chemical modification. Example 3 herein illustrates a chemically modified IFN conl comprised~of an IFN conl moiety connected at the N-terminus to a polyethylene glycol moiety (PEG 12000).
In another aspect, the present methods yield pegylated proteins where the polyethylene glycol moiety is directly attached to a protein moiety, and a separate linking group is absent and no toxic by-products are present. The examples include G-CSF and consensus interferon as described herein. For a population of pegylated G-CSF protein molecules wherein the polyethylene glycol moiety is directly attached to the G-CSF protein moiety (not necessarily a population of N-terminally pegylated G-CSF molecules), one may perform the above reductive alkylation with or without an acidic pH.
In yet another aspect of the present invention, provided are pharmaceLtical compositions of the above. Such pharmaceutical compositions may be for administration for injection, or for oral, pulmonary, nasal or other forms of administration. In general, comprehended by the invention are pharmaceutical compositions comprising effective amounts of monopolymer/protein conjugate products of the invention together with pharmaceutically acceptable diluents, preservatives, solubilizers, emulsifiers, adjuvants and/or carriers. Such compositions include diluents of various buffer content (e. g., Tris-HC1, acetate, i phosphate), pH and ionic strength; additives such as detergents and solubilizing agents (e.g., Tweeri 80, Polysorbate 80), anti-oxidants (e. g., ascorbic acid, sodium metabisulfite), preservatives (e. g., Thimersol,~
benzyl alcohol) and bulking substances (e. g., lactose, mannitol); incorporation of the material into particulate preparations of polymeric compounds such as polylactic acid, polyglycolic acid, etc. or into liposomes. Such compositions may influence the physical state, stability, rate of ~ VlVb release, and rate of y~ Y'wc clearance of the present N-terminally chemically modified proteins. ~, ea., Remington's Pharmaceutical Sciences, 18th Ed. (1990, Mack Publishing Co., Easton, PA 18042) pages 1435-1712, In yet another aspect of the present invention, methods of treatment and manufacture of a medicament are provided. Conditions alleviated or modulated by the administration of the present polymer/G-CSF conjugates (or analogs having the hematopoietic biological properties of naturally occurring G-CSF) are typically those characterized by a reduced hematopoietic or immune function, and, more specifically, a reduced neutrophil count. Such conditions may be induced as a course of therapy for other purposes, such as chemotherapy or radiation therapy. Such conditions may result from infectious disease, such as bacterial, viral-, fungal or other infectious disease. Far example, sepsis results from bacterial infection. Or, such condition may be hereditary or environmentally caused, such as severe chronicweutropenia or leukemias. Age may also play a factor, as in the geriatric setting, patients may have a reduced neutrophil count yr reduced neutrophil mobilization. Some of such conditions are reviewed in * Trademark Filgrastim (r-met Hu G-CSF) in Clinical Practice, Morstyn, G. and T.M. Dexter, eds., Marcel Dekker, Inc., N.Y., N.Y. (1993), 351 pp. Other less-studied conditions which may be alleviated or modulated by administration of the present polymer/G-CSF conjugates may include the reduction of lipids (or cholesterol) in the bloodstream, and certain cardiovascular conditions, as G-CSF may induce production of plasminogen activators. The mode of action of G-CSF (or analogs) in these settings is not well understood at present. The addition of a water soluble polymer; such as polyethylene glycol, may provide practical patient benefits in that the sustained duration of biological activity may allow for fewer G-CSF injections per course of treatment.
Generally, conditions which may be alleviated or modulated by administration of the present polymer/consensus interferon are those to which consensus interferon is applicable and include cell proliferation disorders, viral infections, and autoimmune disorders such as multiple sclerosis.
McManus Balmer, DICP, The Annals of Pharmacotherapy ~Q: 761-767 (1990)(Clinical use of biologic response modifiers in cancer treatment: an overview. Part I. The Interferons). Methods and compositions for the treatment of cell proliferation disorders using consensus interferon are described in PCT WO 92/06707, published April 30, 1992, which is herein incorporated by reference. For example, hepatitis (A; B, C, D, E) may be treatable using the present pegylated consensus interferon molecules. The working example below demonstrates that, ~ v~,tro, chemically modified consensus interferon has 20~ of the biological activity of non-chemically modified consensus interferon.

For all of the above molecules, as further studies are conducted, information will emerge regarding appropriate dosage levels for treatment of various conditions in various patients, and the ordinary skilled worker, considering the therapeutic context, age and general health of the recipient, will be able to ascertain proper dosing. Generally, for injection or infusion, dosage will be between 0.01 ~1g/kg body weight, (calculating the mass of the protein alone, without chemical modification), and 100 ~tg/kg (based on the s ame ) .
The below examples illustrate the various aspects discussed above. In Example 1, the advantages of N-terminally pegylated G-CSF are demonstrated as compared to G-CSF monopegylated at lysine-35 or lysine 41 (of the G-CSF met + 174 amino acid version). Example 2 illustrates the present reductive alkylation in N-terminally pegylating G-CSF. The method provides for a substantially homogenous preparation of N-terminally pegylated G-CSF. Example 3 illustrates the present reductive alkylation in~N-terminally pegylating consensus interferon.
A. preps at s on o~Recombinant human met-G-CSF
Recombinant human met-G-CSF (referred to as "rhG-CSF" or "r-met-hu-G-CSF" from time to time herein) was prepared as described above according to methods in the Souza patent, U.S. Pat. No., 4,810,643.
The rhG-CSF employed was an E",, coli derived recombinant expression product having the amino acid sequence (encoded by the DNA

j sequence) shown below (Seq.ID NOs. 1 and 2):
ATG ACT CCA TTA GGT CCT GCT TCT TCT CTG CCG CAA AGC TTT CTG
M T P L G P A S S L P Q S F L
CTG AAA TGT CTG GAA CAG GTT CGT AAA ATC CAG GGT GAC GGT GCT
L K C L E Q V R K I Q G D G A
GCA CTG CAA GAA AAA CTG TGC GCT ACT TAC AAA CTG TGC CAT CCG

GAA GAG CTG GTA CTG CTG GGT CAT TCT CTT GGG ATC CCG TGG GCT
E E L V L L G H S L G I P W A
CCG CTG TCT TCT TGT CCA TCT CAA GCT CTT CAG CTG GCT GGT TGT
P L S S C . P S Q A L Q L A G C
CTG TCT CAA CTG CAT TCT GGT CTG TTC CTG TAT CAG GGT CTT CTG
L S Q L H S G L F L Y Q G L L
CAA GCT CTG GAA GGT ATC TCT CCG GAA CTG GGT CCG ACT CTG GAC
Q A L E G I S P E L G P T L D
ACT CTG CAG CTA GAT GTA GCT GAC TTT GCT ACT ACT ATT TGG CAA
T L Q L D V A D F A T T I W Q
CAG ATG GAA GAG CTC GGT ATG GCA CCA GCT CTG CAA CCG ACT CAA
Q M E E L G M A P A L Q P T Q

G A M P A F A S A F Q R R A G
GGT GTA CTG GTT GCT TCT CAT CTG CAA TCT TTC CTG GAA GTA TCT
G V L V A S H L Q S F L E V S
TAC CGT GTT CTG CGT CAT CTG GCT CAG CCG TAA TAG
Y R V L R H L A Q P
(This was also the non-pegylated composition used for the control animals.) Alternatively one may use purchased Neupogen~ for the following pegylation procedures.
B. ~~'~~_ratinn c~f Pegylated G-CSF
A 10 mg/ml solution of the above rh-G-CSF, in 100 mM Bicine*pH 8.0, was added to solid SCM-MPEG
(N-hydroxy succinimidyl esters of carboxymethyl methoxy polyethylene glycol) (Union Carbide) with an average "' Trademark molecular weight of 6000 Daltons. This gave a 1.5 molar excess of SCM-MPEG to rh-G-CSF. After one hour with gentle stirring, the mixture was diluted to 2 mg/ml with sterile water, and the pH was adjusted to 4.0 with dilute HC1. The reaction was carried out at room temperature. At this stage, the reaction mixture consisted mainly of three forms of mono-pegylated rh-G-CSF, some di-pegylated rh-G-CSF, unmodified rh-G-CSF and reaction bi-product (N-hydroxy succinimide).
C. prPparatinn of N-terminally Peqylated rh-G-CSF
The three forms of monopegylated rh-G-CSF were separated from each other using ion exchange chromatography. The reaction mixture was loaded (l mg protein/ml resin) onto a Pharmacia S Sepharose FF~column (Pharmacia XK50/30 reservoir, bed volume of 440 ml) equilibrated in buffer A (20 mM sodium acetate, pH 4.0).
The column was washed with 3 column volumes of buffer A.
The protein was eluted using a linear gradient from 0-23% buffer B (20 mM sodium acetate, pH 4.0, 1M NaCl) in 15 column volumes. The column was then washed with one column volume of 100% buffer 8 and reequilibrated with 3 column volumes of buffer A. The flow rate for the entire run was maintained at 8 ml/min. The eluent was monitored at 280 nm and 5 ml fractions were collected.
Fractions containing the individual monopegylated species were pooled according to FIGURE lA. These pools were concentrated with a 350 mL Amicon stirred cell using a YM10 76 mm membrane.
Pooled fractions from the ion exchange chromatography were subjected to size exclusion chromatography to separate di-pegylated species from monopegylated species. Typically, 5-10 mg in 2-5 ml of solution were loaded onto a 120 ml Pharmacia Superdex~"
75 HR 16/60 column equilibrated with 20 mM sodium * Tradenaark . - 28 -acetate pH 4Ø The column was run at 1.5 ml/min for 100 min. Two ml fractions were collected. The protein content of the eluent was monitored at 280 nm.
Fractions from separated peaks were pooled and subjected to analysis. The table below compares the proportional yields for each peak.

RP1_~"t-;Ve Yields and Site of Modification Figure lA
Site of Modification Reference Relative Yields N-Terminus Peak lA 3 L sine-35 Peak 2A 2 L sine-41 Peak 3A 1 Under these conditions, the lysines at positions 17 and 24 probably were not significantly pegylated.
D. Characterization Five analyses were done to characterize each sample: (1) SDS-Page (Figure lB), (2) Size exclusion chromatography HPLC ("SEC HPLC")tFigure 2), (3) peptide mapping analysis (Figures 3A, 3B, and 3C),(4) y~ yitro G-CSF bioassay (Figure 4), and (5) jn vivo testing in hamster (Figures 5A and 5B).
With regard to the composition of each sample, results demonstrate that, of the N-terminally monopegylated G-CSF, the samples showed a greater than 95% N-terminally pegylated composition, with the remainder probably being unpegylated material (although the remainder of the samples is lower than the detection limit of the assay). With regard to the percent monopegylated for each of the three types of monopegylated material (N-terminal, pegylated at lysine 35, and pegylated at lysine 41), the N-terminal and the lysine 41 demonstrated greater than 97% monopegylated, _ 29 _ and the lysine 35 pegylated material being somewhat lower, probably due to the instability of the molecule in the assay conditions. To summarize, the following results were obtained:

Percent Composition of N-terminally peaylated G-CSF
Non-Reduced N-Terminal SDS PAGE SEC HPLC Se encin Mono-pegylated G-CSF 97.44 99.43 96.6 Unmodified G-CSF 2.56 0.57 3.4 , * The N-terminal sequencing, as discussed j,~,~ is not here considered quantitative, as there may have been artifactual separation of the polyethylene glycol molecule from the N-terminus of the protein during the sequencing process.

Percent Mono~egylated for Three Species N-terminal LYS35 PEG- LYS41 PEG-GCSE GCSE** PEG-GCSF

RI/W=.96) RI/W=.72) (RI/W=1 .12 *

Non-reduced SDS-PAGE 97.44 77.41 100.00 SEC HPLC 99:43 93.38 99.96 * RI/W refers to the Index of Refraction/Ultraviolet light absorbance ratio, and is used to estimate the number of polyethylene glycol molecules per molecule of protein. It is calculated from the SEC HPLC data using i an Index of Refraction for polyethylene glycol and an ultraviolet absorbance for protein.
** Note that this species is unstable under the assay conditions used.
METHODS
1. SDS-P,~,GE. SDS-PAGE was carried out in a non-reduced 4-20% ISS Daiichi Pure Chemicals, Co., Tokyo, Japan minigel using a Coomassie Brillant Blue R-250 stain. The gel was scanned using a molecular Dynamics Densitometer with Image Quanta:
Results: Results are presented in FIGURE 1B. Lane number 1 (from the left hand side) included molecular weight protein standards (Novex Mark 12 Molecular Weight Standards). Lane 2 contains 3 ~g rh-G-CSF standard.
Lane 3 contains the SCM-PEG-GCSE reaction mix, with 10 ~tg loaded. Lane 4 contains N-terminally monopegylated G-CSF, with 10 )tg loaded. Lane 5 contains 10 ~tg of monopegylated G-CSF with the pegylation site at the lysine found at the 35th residue from the N-terminal methionine. Lane 6 contains 10 ~tg of monopegylated G-CSF with the pegylation site at the lysine found at the 41st residue from the N-terminal methionine. As can be seen, Lane 3, containing the N-terminally monopegylated material, shows a single band 2. Size Exc~~ sio Chromatog;~phy-Hiqh Pressure Lid iru d Ch,~~matoara~hv.. SEC-HPLC was carried out using a Waters HPLC system with a Biosep SEC 3000 column, using 100 mM sodium phosphate, pH 6.9, lml/min for 20 minutes. The signal was monitored at 280 nm.
Results: As can be seen from Figure 2, line "C,"
containing the N-terminally monopegylated rh-G-CSF
contains a single peak, as do lines "D" (Lys-35 monopegylated material) and "E" (Lys-41 monopegylated '" Trademark material). This indicates substantial purity among the separated fractions of monopegylated G-CSF.
3. Pegtide mag8ina. The following methods were used. Three samples, called "Mono-PEG 1", "Mono-PEG-2", and "Mono-PEG-3", were analyzed. (a) Reductive alkylation. 500 ~tg aliquots of mono-PEG G-CSF were speed vac dried and reconstituted to a concentration of 1 mg in 950 X11 in 0.3 M Tris-HCl containing 6 M
Guanidinum HCl and l mM EDTA pH 8.4. Samples were then S-carboxymethylated by adding iodoacetic acid and incubated at 37°C for 20 minutes. Samples were then desalted using Sephadex~G-25 Quick Spin Protein 'Columns and buffer exchanged. After desalting and buffer exchange, sample concentration was adjusted to 0.5 mg/ml using additional buffer. (b) Endoproteinase SV8 digestion. Samples were digested with SV8 (enzyme to substrate, ratio of 1:25) at 25°C for 26 hours . (c) HPLC
peptide mapping. Protein digests were injected onto a .vydac C4*column (4.6 x 250 mm, 5 ~t particle size, 300 pore size) and peptides were mapped by HPLC using a linear gradient of acetonitrile in 0.1% TFA. Peptides were manually collected and dried in a Speed Vac for sequence analysis. Results: As compared to a reference standard, (i) (FIGURE 3A) for "Mono-PEG-1", (the N-terminally mono-pegylated material), a peak at 57.3 minutes diminished and a new peak appeared at 77.5 minutes: (ii) (FIGURE 38) for "Mono-PEG-2", (the lysine pegylated material), there was a decrease in peak height for a peptide With a retention time of 30.3 30 minutes, and a new peak eluted at 66.3 minutes: (iii) (FIGURE 3C) for "Mono-PEG-3" (the lysine 41 pegylated material), the peak at retention time of 30.3 minutes was missing, and a new peak appeared at 66.4 minutes.
These peptides were the only significant differences in 35 the sample maps. There were some small incomplete * Trademark cleavages seen on either side of the peptide at 86.1 minutes due to minor digestion differences. (d) terminal seauence analysis. Each of the "new" peptides in the above maps were N-terminally sequenced for identification. The dried peptides were reconstituted in 0.1% TFA and sequenced on an ABI protein sequencer.
For "Mono-PEG-1" (the N-terminally pegylated material), 60% of the "new" peak (at 77.5 minutes) was sequenced for 10 cycles. The initial yield was less than 5%, indicating that the N-terminal methionyl residue is blocked by a polyethylene glycol molecule. It is noted that this initial peptide should have resulted in a zero initial yield, and the <5% yield observed may be from detachment of the polyethylene glycol from the N-terminal methionyl during sequence analysis. The sequence detected Was that of the N-terminal peptide, M-T-P-L-G-P-A-S-S. For "Mono-PEG-2", (the lysine 35 pegylated material), 80% of the total peak volume was collected for the peak at 66.3 minutes, and was sequenced fox 9 cycles. The recovery of lysine 35 was significantly low, indicating pegylation at position 35.
The recovery of lysine 41 was consistent with the other residue, indicating no modification of this position.
The peptide at 30.3 minutes decreased in peak height compared to the corresponding peak in the standard reference map. The peptide at 30.3 minutes is only 57.5% of the peak area of the corresponding peptide.
The sequence detected for this species was K-L-C-A-T-Y-K-L. For "Mono-PEG-3", the lysine 41 material, 80% of the total peak volume collected for the peptide eluting~at 66.4 minutes was sequenced for 9 cycles. The sequence detected was K-L-C-A-T'Y-K-L, and contained lysine residues 35 and 41. The recovery of lysine 35 was consistent with other residue recoveries.
The recovery of lysine 41 was significantly lower indicating pegylation at position 41. RP~~; "Mono-PEG-1" is N-terminally monopegylated material; "Mono-PEG-2" is lysine 35 partially pegylated; and "Mono-PEG-3" is lysine 41 pegylated material. By comparing both the reference standard (non-pegylated G-CSF) and GCSE.
monopegylated 1, 2, and 3 peptide maps, it was found that both the "Mono-PEG-2" (lysine 35) and "Mono-PEG-3"
(lysine 41) maps exhibit slightly diminished peak heights for the N-terminal peptide. This indicates that the lysine 35 and lysine 41 material contains a small amount of N-terminally pegylated material or that the N-terminal methionine has a small percentage of pegylation.
4. In vitro acts. The material was active. FIGURE 4 illustrates the results of ~ yitro assays. As can be seen, the N-terminally monopegylated material had 68% of the activity of non-modified rhG-CSF.
Methods: The G-CSF iI1 vitro bioassay is a mitogenic assay utilizing a G-CSF dependent clone of murine 32D
cells. Cells were maintained in Iscoves medium containing 5% FBS and 20 ng/ml rhG-CSF. Prior to sample addition, cells were prepared by rinsing twice with growth medium lacking rhG-CSF. An extended twelve point rhG-CSF standard curve was prepared, ranging from 48 to .Sng/ml (equivalent to 4800 to 50 IU/ml). Four dilutions, estimated to fall within the linear portion of the standard curve, (1000 to 3000 IU/ml), were prepared for each sample and run in triplicate. Because of their apparent lower activity ~ vitro, the pegylated rhG-CSF samples were diluted approximately 4-10 times less. A volume of 401 of each dilution of sample or standard is added to appropriate wells of a 96 well microtiter plate containing 10,000 cells/well. After forty-eight hours at 37°C and 5:5% C02, 0.5~tmCi of , methyl-3H-thyrnidine was added to each well. Eighteen hours later, the plates were then harvested and counted.
A dose response curve (log rhG-CSF concentration vs.
CPM-background) was generated and linear regression analysis of points which fall in the linear portion of the standard curve was performed. Concentrations of unknown test samples were determined using the resulting linear equation and correction for the dilution factor.
Results: Results are presented in FIGURE 4. As can be seen, of the three monopegylated species, N-terminally monopegylated G-CSF demonstrates the highest i~1 vitro biological activity.
5. Tn vivo a~tivitv. ~ vivo testing confirmed the activity of the N-terminally pegylated material. The is vivo testing was carried out by dosing male golden hamsters with a 0.1 mg/kg of sample, using a single subcutaneous injection. Four animals were subjected to terminal bleeds per group per time point.
Serum samples were subject to a complete blood count on the same day that the samples were collected. The average white blood cell counts were calculated. As can be seen in FIGURES 5A and 5B, the response from each material peaks after one day following a single subcutaneous injection of 0.1 mg/kg. Two of the monopegylated materials, (N-terminal and Lys-35) showed prolonged responses, while the response for the protein pegylated at lysine-41 showed no increase in is vivo ' activity over unmodified rhG-CSF (indeed it shows less, FIGURE 58). These results illustrate that attaching a single polyethylene glycol molecule can dramatically alter the therapeutic profile of a protein and that the benefit of pegylating a protein can be dependent upon the site of modification. (The net average WBC area under the curve after the single subcutaneous injection (calculated according to CRC Standard Mathematical S

Tables, 26th Ed. (Beyer, W.H., Ed.) CRC Press Inc., Boca Baton, FL 1981. p. 125) was similar for the Lys-35 and N-terminal monopegylated species.) E. Stability Studies In addition, stability studies were performed on the N-terminal and Lys-35 monopegylated species as prepared above. (The Lys-41 material was not used as it demonstrated no additional activity beyond unmodified G-CSF). These studies demonstrate that the N-terminally pegylated G-CSF is unexpectedly more stable upon storage than the other form of monopegylated G-CSF, monopegylated lysine 35. Stability was assessed in terms of breakdown of product, as visualized using SEC-HPLC.
Methods: N-terminally pegylated G-CSF and lysine-35 monopegylated G-CSF were studied in two pH levels, pH 4.0 and pH 6.0 at 4°C, each for up to 16 days.
Elevating the pH to 6.0 provides an environment for accelerated stability assays. For the pH 6.0 samples, N-terminal monopegylated G-CSF and Lysine 35 monopegylated G-CSF as prepared above were placed in a buffer containing 20 mM sodium phosphate, 5 mM sodium acetate, 2.5 % mannitol, 0.005 % Tween-80, pH 6.0 at a final protein concentration of 0.25 mg/ml. One ml aliquots were stored in 3 ml sterile injection vials.
Vials of each was stored at 4°C and 29°C for up to 16 days. Stability was assessed by SEC-HPLC tracings.
If the later measurements stayed the same (as ascertained by visual inspection) as the initial (Time =
0) measurements, the sample was considered to be stable for that length of time.
Results: Results are illustrated in FIGURES 6A-6C.
(a) Comparison at pH 6.0 at 4°C. FIGURE 6A shows the 4°C SEC=HPLC profiles for N-terminally monopegylated * Trademark G-CSF at pH 6 over time and FIGURE 6B shows the 4°C
SEC-HPLC profiles for lysine-35 monopegylated G-CSF at pH 6 over time. One interpretation is that the Lys-35 material is breaking down to a material with a molecular weight similar to that of unmodified G-CSF.
(b) Extended duration at pH 4.0 at 4°C. PH 4.0 and 4°C
provides something of a control illustrating relatively stable conditions in that the N-terminal species shows no degradation. For the Lys 35 species, the break down of the material is still occurring, but at a much slower rate.
(c) Comparison at pH 6.0 at 4°C. FIGURES 6C illustrates the SEC-HPLC profiles for the monopegylated G-CSF's under these conditions, under extended time periods. As can be seen, at pH 6.0 and 4°C, the lysine-35 material exhibits no increase in depegylation at day 16 or day 35 beyond what was seen for day 6 (FIGURE 6B). This indicates that depegylation (instability) does not change, under those conditions, beyond day 6.
This example demonstrates a method of preparing a substantially homogenous population of monopegylated G-CSF using reductive alkylation, and characterization of this population. Recombinant G-CSF
as described in the above example was used. As can be seen, not only do the present methods provide advantages in terms of yield of N-terminally chemically modified material, but also, the amine linkages of the present reductive alkylation process produce substantially more stable products as demonstrated by a large difference in the degree of aggregation upon storage.

.l A. ~r ~a_rat,'_on of the 'mono-methoxvnolyeth_,y1_ene ,~1 ~,r~,n1_-GCSF conjugates G~i h h ai of a tac ent at the N-terminal"g-amino residue.
To a cooled (9 oC),stirred solution of rhG-CSF
(1 ml, 5 mg/ml as described in the Example above) in 100 mM sodium phosphate, pH 5, containing 20 mM NaCNBH3, was added a 5-fold molar excess of methoxypolyethylene glycol aldehyde (MPEG)(average molecular weight, 6 kDa).
The stirring of the reaction mixture was continued at the same temperature.
The extent of the protein modification during the course of. the reaction was monitored by SEC HPLC
using Bio-Sil*SEC 250-5*column (BIO-RAD) eluted with 0.05 M NaH2P04,0.05 M Na2HP04,0.15 M NaCl, 0.01 M NaN3, pH 6.8 at 1 ml/min.
After 10 hours the SEC HPLC analysis indicated that 92% of the protein has been converted to the mono-MPEG-GCSE derivative. This can be seen in FIGURE 7, which is a recording of the protein concentration (as determined by absorbance at A2g0) and shows the peak eluting at 8.72 minutes as monopegylated G-CSF, and a minor peak of unreacted G-CSF eluting at 9.78 minutes.
As a comparison, FIGURE 8 shows the peaks obtained when using N-hydroxysuccinimidyl ester of MPEG.
The molecular weight Was also 6kDa. As can be seen, the mixture obtained from this reaction was: tri-MPEG-GCSF
conjugated (shoulder at approximately 7.25 minutes), di-MPEG-GCSE conjugate (peak at 7.62 minutes), mono-MPEG-GCSF conjugate (peak at 8.43 minutes) and unreacted G-CSF (peak at 9.87 minutes).
At this 10 hour time point, where 92% of the protein had been converted to monopegylated material, the pH of the reaction mixture was adjusted to pH 4 with 100 mM HC1 and the reaction mixture was diluted 5 times with 1 mM HC1.
Tradern~rk The mono~MPEG-GCSE derivative was purified by ion exchange chromatography using HiLoad 16/10 S
Sepharose HP column (Pharmacia) equilibrated with 20 mM
sodium acetate buffer, pH 4. The reaction mixture was loaded on the column at a flow rate of 1 ml/min and the unreacted MPEG aldehyde eluted with three column volumes of the same buffer. Then a linear 400 minute gradient from 0% to 45% 20 mM sodium acetate, pH 4, containing 1 M NaCl was used to the elute the protein-polymer conjugate-at 4oC.
Fractions containing the mono-MPEG-GCSF
derivative were pooled, concentrated and sterile filtered.
Various mono-MPEG-GCSF conjugates obtained by modifying rh-G-CSF with MPEG aldehydes of different average molecular weight (12, 20 and 25 kDa) were prepared in a similar manner.
B. Analysis of ~popeqvlated G-CSF
1. Molecular Weight The molecular weight at the monopegylated conjugates was determined by SDS-PAGE, gel filtration, matrix assisted laser desorption mass spectrometry, and equilibrium centrifugation. These results are presented in Table 4, below.
"' Trademark t~tn~~ W~~hts of N-terminaW Y Alkvlated Mono-I~E(,~-GCSF Cone" a Conjugate Md MW gel tai ma~~ ICI ultra-~atimated filtrationapsct~ertrycantrituQation S ~a -se ~A ~ a.~s~~~
~

t9pFx-(6kDa)- 24800 53024 24737 25548 OCSF

hJPEG-(l2kDa)- 30800 124343 30703 29711 MpE~
(20kDa)- 38800 221876 38577 38196 CaC.SF
(25kDa)- 43800 333266 N/D N/D
The structure of the prepared N-terminal mono-MPEG-GCSF conjugates was confirmed using the methods of N-terminal protein sequencing and peptide mapping. Cyanogen bromide cleavage of the N-terminal methionyl residue resulted in removal of the polyethylene glycol.
2. ~iologyal ~tivitv The y~ iv tro biological activity of the pegylated I~EG-GCSF conjugates'was determined by measuring the stimulated uptake of 3H.thymidine into mouse bone marrow cells.
The in vivo biological activity was determined by subcutaneous injection to hamsters MPEG-GCSF
conjugates or rhG-CSF (at 100mg/kg) and measuring total white blood cell count. Bioactivity as compared to non-derivatized G-CSF was calculated as the area under i the WBC/time curve after subtracting the vehicle control curve. Relative bioactivity of the MPEG-GCSF derivatives was expressed as the percentage bioactivity compared to unmodified G-CSF.
This is illustrated in FIGURE 9, which is a graph illustrating the total white blood cell response to mono-N-terminal I~EG-GCSE conjugates prepared by reductive alkylation of rhG-CF with MPEG aldehydes of different molecular weights (6kDa, l2kDa, and 20kDa).
As can be~seen, all monopegylated molecules elicited a response. The higher the molecular weight of the polyethylene glycol moiety used, the higher the white blood cell count achieved, except the l2kDa achieved a slightly higher count than did the 20kDa version at day 2.
3. Stability Studies N-terminally pegylated G-CSF's prepared by the two different chemistries (amide vs. amine of the reductive alkylation here) were compared for the degree of aggregation. Unexpectedly, N-terminally pegylated G-CSF using the amine chemistry was found to be substantially more stable than N-terminally pegylated G-CSF with an amide linkage (NHS chemistry as described in Example 1).
Methods: Both N-terminally pegylated G-CSF
samples.were in 10 mM NaOac pH4.0 with 5% sorbitol, at a concentration of lmg protein/ml.. The G-CSF's were pegylated with PEG 6000 for each. The amide-linked conjugate was prepared as in Example 1, and the amine linked conjugate was prepared as in Example 2. Six samples of each were stored for eight weeks at 45°C. At the end of eight weeks, the degree of aggregation was determined using size exclusion chromatography and ion exchange chromatography.

Results: The results demonstrate that the present reductive alkylation methodis advantageous over aceylation because, surprisingly, it produces a material with far fewer aggregates after 8 weeks at elevated temperatures. The table below shows the percent of non-aggregated material ("main peak" material) for both materials using size exclusion chromatography (SEC) or ion exchange (IE):
l0 ~ TABLE 5 Sample:8 wks, 45C % Main Peak SEC/IE

Amine 82%/84%

Amide 37%/65%*

* This is relatively high because ion exchange does not allow for full analysis of the aggregation.
E~ LE 3 This example demonstrates chemically modified consensus interferon. More specifically, this example demonstrates a method of preparing a substantially homogenous population of monopegylated IFN-cons, and characterization of this population.
It should be noted that while the,present example uses IFN-conl, any of the consensus interferons as set forth above may be chemically~modified. Such chemical modification may be with any of the Water soluble polymers as listed above, although PEG is used here. For pegylation, PEG 12000 is used here, although any water soluble PEG species may be used (PEG 12000 was selected for ease. in handling and convenience). Again, a variety of means for chemical modification are available (such as acetylation) but, for selective N-terminal chemical modification, such as N-terminal pegylation, the present reductive alkylation method as described in this example is preferred.
A. Pr~~aration of Consensy,S TBtPr~
IFN-aconl (here referred to as IFN-conl) as described in Figure 2 of U.S. Patent No. 4,695,623, was used for the preparation of monopegylated consensus IO interferon. The IFN-conl was produced by expression of exogenous DNA in bacteria, and contained a methionyl residue at the N-terminus.
B. Pegy a s o~,of Consensug Interferon I5 To a cooled (4 oC), stirred solution of IFN-conl (3.45 mg/ml, containing 35.25% of the N-terminally blocked form) in 100 mM sodium phosphate, pH 4.0;
containing 20 mM NaCN8H3 was added a 8-fold molar excess of methoxypolyethylene glycol aldehyde (1~EG)(average 20 molecular weight 12 kDa).
The extent of the protein modification during the course of the reaction was monitored by reverse phase HPLC using a polymer-based poly(styrene/divinylbenzene) column, such as PLRP -S (PL
25 Separation Sciences Polymer Laboratories).
After 10 hours the reverse phase HPLC analysis indicated that 80% of the protein with unblocked a-amino group at the N-terminus has been converted to the N~EG-IFN-conl derivative.
30 At the 10 hour time point, the reaction mixture was diluted S times with water and the mono-MPEG-IFN-Conl derivative was purified by ion exchange chromatography using HiLoad 16/10 S Sepharose*
HP column (Pharmacia) equilibrated with 20 mM sodium 35 acetate buffer, pH 4Ø The reaction mixture was loaded * Trademark l ) on the column at a flow rate of I ml/min and the unreacted MPEG aldehyde eluted with three column volumes of the same buffer. Then a linear 420 minute gradient from 0% to 75% of 20 mM sodium acetate, pH 4.0, containing 1 M NaCl was used to the elute the protein-polymer conjugate at 4oC.
Fractions containing the. mono-MPEG-IFN-Conl derivative Were pooled, concentrated and sterile filtered.
C. Analys~,g, o~ ono~egvlaltP~ GonE,ensLS interferon I. HomoAeneitv The homogeneity of the purified mono-MPEG-IFN-Conl conjugates was determined by SDS-PAGE
using 10-20% or 4-20% precast gradient gels (Integrated Separation Systems). The gels showed a main band at MW
35 kDa.
To characterize the effective size (hydrodynamic radius) of each mono-1~EG-IFN-conk species a Superose 6 HR 10/30 (Pharmacia) gel filtration column was used. Proteins were detected by W absorbance at 280 nm. The BIO-RAD gel filtration standards served as globular protein molecular weight markers.
The structure of the purified N-terminal mono-MPEG-IFN-conl conjugates was confirmed using the methods of N-terminal protein sequencing and peptide mapping. ~ ' It is noted that this IFN-conl preparation contained some N-terminally blocked material, and this 34 material was not pegylated. The material which was pegylated, however, was monopegylated at the N-terminus.
Thus, in this type of situation, one may wish to use other means to separate the blocked from the unblocked material, such as ion exchange or size exclusion chromatography.
* Trademark 2. ~,ological Activity The yn vitro biological activity of the mono-1~EG-IFN Conl conjugates was determined by measuring their antiviral bioactivity. The ,yn vitro biological activity of the mono-I~EG-IFN-Conl conjugates was determined by measuring their antiviral bioactivity in human (HeLa) cells.
It was found that the mono-1~EG (12 kDa)-IFN-Conl conjugate shows 20% ya vitro bioactivity tin U/mg of protein) when compared to the unmodified species. As noted above for pegylated G-CSF, the j,n vitro assays, while useful to demonstrate biological activity, may show a rather low level of activity for chemically modified proteins because of characteristic sustained release. The ~ vivo biological activity may be higher than the ~ ~,~itro biological activity.
D. Gh~"mi cal 1 y~difi a~~ consen~,ls inte~,~~,,ron w h The present reductive alkylation was also performed on the above IFN-conl which had the portion of N-terminally blocked molecules pre-removed. Both PEG
12000 and PEG 20000 were used in the reductive alkylation method as described above.
The molecular apparent molecular weights were as follow:
Apparent MW by Apparent MW by Con'u ate Gel Filtration SDS-PAGE

monoMPEG(l2kDa) 104.0 kDa 35.6 kDa IFN-con monoMPEG120kDa) 175.1 kDa 55.4 kDa IFN-con Analysis of the IFN-conl 20 kDa PEG conjugate using FPLC ion exchange chromatography resulted in three peaks:
MonoMPEG-IFN-conl; 66% of the total area (eluting at 265.93 ml) Protein aggregate and oligo MPEG-IFN-conl conjugate: 24% of the total area (eluting at 238.42 mI ) ; and Unreacted IFN-conk 10% of the total area (eluting at 328.77 ml).
The conditions were not further optimized. One may further separate the monopegylated material using chromatographic or other methods.
I5 While the present invention has been described in terms of preferred embodiments, it is understood that variations and modifications will occur to those skilled in the art. Therefore, it is intended that the appended claims cover all such equivalent variations which come within the scope of the 'invention as claimed.

i ) - ~46 - .
SEQUENCE LISTING
(1) GENERAL INFORMATION:
(i) APPLICANT: AMGEN INC.
(ii) TITLE OF INVENTION: N-Terminally Chemically Modified Protein Composition and Methods (iii) NUMBER OF SEQUENCES: 2 (iv) CORRESPONDENCE ADDRESS:
(A) ADDRESSEE: Amgen Inc.
(B) STREET: 1846 Dehavilland Drive (C) CITY: Thousand Oaks (D) STATE: California (E) COUNTRY: USA
(F) ZIP: 91320 (v) COMPUTER READABLE FORM:
(A) MEDIUM TYPE; Floppy disk (B) COMPUTER: IBM PC compatible (C) OPERATING SYSTEM: PC-DOS/MS-DOS
(D) SOFTWARE: PatentIn Release X1.0, Version X1.25 (vi) CURRENT APPLICATION DATA:
(A) APPLICATION NUMBER:
(B) FILING DATE:
(C) CLASSIFICATION:
(viii) ATTORNEY/AGENT INFORMATION:
(A) NAME: Pessin, Karol M.
(C) REFERENCE/DOCKET NUMBER: A-286 (2) INFORMATION FOR SEQ ID N0:1:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 531 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA

(xi) SEQUENCE DESCRIPTION: SEQ ID NO:1:

(2) INFORMATION FOR SEQ ID N0:2:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 175 amino acids (8) TYPE: amino acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (ii) MOLECULE TYPE: protein (xi) SEQUENCE DESCRIPTION: SEQ ID N0:2:
Met Thr Pro Leu Gly Pro Ala Ser Ser Leu Pro Gln Ser Phe Leu Leu Lys Cys Leu Glu Gln Val Arg Lys Ile Gln Gly Asp Gly Ala Ala Leu Gln Glu Lys Leu Cys Ala Thr Tyr Lys Leu Cys His Pro Glu Glu Leu 35 40 . 45 Val Leu Leu Gly His Ser Leu Gly Ile Pro Trp Ala Pro Leu Ser Ser Cys Pro Ser Gln Ala Leu Gln Leu Ala Gly Cys Leu Ser Gln Leu His Ser Gly Leu Phe Leu Tyz Gln Gly Leu Leu Gln Ala Leu Glu Gly Ile Ser Pro Glu Leu Gly Pro Thr Leu Asp Thr Leu Gln Leu Asp Val Ala Asp Phe Ala Thr Thr Ile Trp Gln G1n Met Glu Glu Leu Gly Met Ala Pro Ala Leu Gln Pro Thr Gln Gly Ala Met Pro Ala Phe Ala Ser Ala Phe Gln Arg Arg Ala Gly Gly Val Leu Val Ala Ser His Leu Gln Ser Phe Leu Glu Val Ser Tyr Arg Val Leu Arg His Leu Ala Gln Pro

Claims (6)

1. A substantially homogeneous preparation of a pegylated protein comprising polyethylene glycol conjugated to the protein via an amino group on the protein, wherein the protein is monopegylated and the polyethylene glycol is conjugated predominantly at the N-terminus of the protein.
2. A substantially homogeneous preparation of a peglyated protein comprising polyethylene glycol conjugated to the protein via an amino group on the protein, wherein the protein is monopegylated and the polyethylene glycol having a-molecular weight of between about 2 kDA and 100 kDa is conjugated predominantly at the N-terminus of the protein.
3. A pharmaceutical composition comprising (a) the preparation of claim 1 or 2, and (b) a pharmaceutically acceptable diluent, carrier or adjuvant.
4. A method of treating a hematopoietic disorder comprising administering to a patient in need thereof a preparation of claim 1, a preparation of claim 2 or a composition of claim 3.
5. A use of a preparation of claim 1, a preparation of claim 2 or a composition of claim 3 for treating a hematopoietic disorder in a patient in need thereof.
6. A use of a preparation of claim 1, a preparation of claim 2 or a composition of claim 3 for the production of a medicament for treating a hematopoietic disorder in a patient in need thereof.
CA002472085A 1994-10-12 1995-02-08 N-terminally chemically modified protein compositions and methods Abandoned CA2472085A1 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US08/321,510 US5824784A (en) 1994-10-12 1994-10-12 N-terminally chemically modified protein compositions and methods
US08/321,510 1994-10-12
CA2307142A CA2307142C (en) 1994-10-12 1995-02-08 N-terminally chemically modified protein compositions and methods

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
CA2307142A Division CA2307142C (en) 1994-10-12 1995-02-08 N-terminally chemically modified protein compositions and methods

Publications (1)

Publication Number Publication Date
CA2472085A1 true CA2472085A1 (en) 1996-04-25

Family

ID=23250903

Family Applications (3)

Application Number Title Priority Date Filing Date
CA002178752A Expired - Lifetime CA2178752C (en) 1994-10-12 1995-02-08 N-terminally chemically modified protein compositions and methods
CA2307142A Expired - Lifetime CA2307142C (en) 1994-10-12 1995-02-08 N-terminally chemically modified protein compositions and methods
CA002472085A Abandoned CA2472085A1 (en) 1994-10-12 1995-02-08 N-terminally chemically modified protein compositions and methods

Family Applications Before (2)

Application Number Title Priority Date Filing Date
CA002178752A Expired - Lifetime CA2178752C (en) 1994-10-12 1995-02-08 N-terminally chemically modified protein compositions and methods
CA2307142A Expired - Lifetime CA2307142C (en) 1994-10-12 1995-02-08 N-terminally chemically modified protein compositions and methods

Country Status (20)

Country Link
US (7) US5824784A (en)
EP (5) EP1564219A1 (en)
JP (8) JP3177251B2 (en)
KR (2) KR100248111B1 (en)
CN (4) CN1229388C (en)
AT (2) ATE277078T1 (en)
CA (3) CA2178752C (en)
DE (3) DE69533556T2 (en)
DK (1) DK0733067T3 (en)
ES (2) ES2131811T3 (en)
GR (1) GR3030526T3 (en)
HK (2) HK1008787A1 (en)
IL (3) IL112585A (en)
LU (1) LU91006I2 (en)
MX (1) MX9602259A (en)
NL (1) NL300106I2 (en)
NZ (1) NZ281469A (en)
PT (1) PT822199E (en)
WO (1) WO1996011953A1 (en)
ZA (1) ZA951008B (en)

Families Citing this family (549)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE68925966T2 (en) * 1988-12-22 1996-08-29 Kirin Amgen Inc CHEMICALLY MODIFIED GRANULOCYTE COLONY EXCITING FACTOR
US20020177688A1 (en) * 1988-12-22 2002-11-28 Kirin-Amgen, Inc., Chemically-modified G-CSF
US5581476A (en) * 1993-01-28 1996-12-03 Amgen Inc. Computer-based methods and articles of manufacture for preparing G-CSF analogs
US5951974A (en) * 1993-11-10 1999-09-14 Enzon, Inc. Interferon polymer conjugates
WO1995021629A1 (en) * 1994-02-08 1995-08-17 Amgen Inc. Oral delivery of chemically modified proteins
US6001968A (en) 1994-08-17 1999-12-14 The Rockefeller University OB polypeptides, modified forms and compositions
US6429290B1 (en) 1994-08-17 2002-08-06 The Rockefeller University OB polypeptides, modified forms and derivatives
US20030053982A1 (en) * 1994-09-26 2003-03-20 Kinstler Olaf B. N-terminally chemically modified protein compositions and methods
US5824784A (en) 1994-10-12 1998-10-20 Amgen Inc. N-terminally chemically modified protein compositions and methods
WO1996041813A2 (en) * 1994-11-09 1996-12-27 Offord Robin E Functionalized polymers for site-specific attachment
US6008202A (en) * 1995-01-23 1999-12-28 University Of Pittsburgh Stable lipid-comprising drug delivery complexes and methods for their production
US5795587A (en) 1995-01-23 1998-08-18 University Of Pittsburgh Stable lipid-comprising drug delivery complexes and methods for their production
DE69632062T2 (en) 1995-11-02 2004-11-18 Schering Corp. CONTINUOUS, LOW-DOSE CYTOKINE INFUSION THERAPY
US5908621A (en) 1995-11-02 1999-06-01 Schering Corporation Polyethylene glycol modified interferon therapy
TW517067B (en) * 1996-05-31 2003-01-11 Hoffmann La Roche Interferon conjugates
EP2002846B1 (en) 1996-12-06 2017-01-25 Amgen Inc. Combination therapy using an IL-1 inhibitor for treating IL-1 mediated diseases
WO1998032466A1 (en) * 1997-01-29 1998-07-30 Polymasc Pharmaceuticals Plc Pegylation process
RO121474B1 (en) 1997-06-06 2007-06-29 Kyowa Hakko Kogyo Co., Ltd. Chemically modified polypeptides
US6753165B1 (en) * 1999-01-14 2004-06-22 Bolder Biotechnology, Inc. Methods for making proteins containing free cysteine residues
EP1881005B1 (en) * 1997-07-14 2013-04-03 Bolder Biotechnology, Inc. Derivatives of G-CSF and related proteins
US20080076706A1 (en) 1997-07-14 2008-03-27 Bolder Biotechnology, Inc. Derivatives of Growth Hormone and Related Proteins, and Methods of Use Thereof
US6017876A (en) * 1997-08-15 2000-01-25 Amgen Inc. Chemical modification of granulocyte-colony stimulating factor (G-CSF) bioactivity
US5985263A (en) * 1997-12-19 1999-11-16 Enzon, Inc. Substantially pure histidine-linked protein polymer conjugates
US5981709A (en) * 1997-12-19 1999-11-09 Enzon, Inc. α-interferon-polymer-conjugates having enhanced biological activity and methods of preparing the same
ATE365563T1 (en) * 1998-04-28 2007-07-15 Applied Research Systems METHOD FOR STEPLY BINDING POLYETHYLENE GLYCOL (PEG) TO POLYPEPTIDES
WO1999059621A1 (en) * 1998-05-15 1999-11-25 Schering Corporation Combination therapy comprising ribavirin and interferon alpha in antiviral treatment naive patients having g chronic hepatitis c infection
NL1009601C2 (en) * 1998-07-09 2000-01-11 Univ Utrecht Compound which can inhibit the binding of a protein to mast cells, use of the compound for the preparation of a medicament, a pharmaceutical preparation, a method of diagnosing a disease and a selection method.
MXPA01000481A (en) * 1998-07-16 2002-11-29 Hyseq Inc Methods and materials relating to novel cd39-like polypeptides.
US6783965B1 (en) 2000-02-10 2004-08-31 Mountain View Pharmaceuticals, Inc. Aggregate-free urate oxidase for preparation of non-immunogenic polymer conjugates
DK2316475T3 (en) 1998-08-06 2017-11-20 Mountain View Pharmaceuticals Inc Isolated tetrameric uricase
US6420339B1 (en) 1998-10-14 2002-07-16 Amgen Inc. Site-directed dual pegylation of proteins for improved bioactivity and biocompatibility
IL142350A0 (en) 1998-10-16 2002-03-10 Biogen Inc Interferon-beta fusion proteins and pharmaceutical compositions containing the same
DK1656952T3 (en) * 1998-10-16 2014-01-20 Biogen Idec Inc Polyalkylene glycol conjugates of interferon beta-1A and applications thereof
US6660843B1 (en) 1998-10-23 2003-12-09 Amgen Inc. Modified peptides as therapeutic agents
DE69934425T2 (en) 1998-10-23 2007-09-27 Amgen Inc., Thousand Oaks THROMBOPOIETIN SUBSTITUTE
US7488590B2 (en) 1998-10-23 2009-02-10 Amgen Inc. Modified peptides as therapeutic agents
JP2002531089A (en) * 1998-11-30 2002-09-24 イーライ・リリー・アンド・カンパニー Erythropoietic compounds
US8288126B2 (en) 1999-01-14 2012-10-16 Bolder Biotechnology, Inc. Methods for making proteins containing free cysteine residues
ATE526401T1 (en) 1999-01-14 2011-10-15 Bolder Biotechnology Inc METHOD FOR PRODUCING PROTEINS WITH FREE CYSTEINE RESIDUE
PT1157037E (en) * 1999-01-29 2003-12-31 Hoffmann La Roche GCSF CONJUGATES
EP1369429A1 (en) * 1999-01-29 2003-12-10 F. Hoffmann-La Roche Ag GCSF conjugates
WO2000051626A1 (en) * 1999-03-01 2000-09-08 Kyowa Hakko Kogyo Co., Ltd. Chemically modified g-csf preparations
US6485718B1 (en) * 1999-04-13 2002-11-26 Pharmacia Corporation Site specific ligation of proteins to synthetic particles
US6924264B1 (en) * 1999-04-30 2005-08-02 Amylin Pharmaceuticals, Inc. Modified exendins and exendin agonists
US8106098B2 (en) * 1999-08-09 2012-01-31 The General Hospital Corporation Protein conjugates with a water-soluble biocompatible, biodegradable polymer
US7459540B1 (en) 1999-09-07 2008-12-02 Amgen Inc. Fibroblast growth factor-like polypeptides
US6348558B1 (en) 1999-12-10 2002-02-19 Shearwater Corporation Hydrolytically degradable polymers and hydrogels made therefrom
US7074878B1 (en) * 1999-12-10 2006-07-11 Harris J Milton Hydrolytically degradable polymers and hydrogels made therefrom
US6831158B2 (en) * 2000-01-10 2004-12-14 Maxygen Holdings Ltd. G-CSF conjugates
US6646110B2 (en) * 2000-01-10 2003-11-11 Maxygen Holdings Ltd. G-CSF polypeptides and conjugates
US6555660B2 (en) * 2000-01-10 2003-04-29 Maxygen Holdings Ltd. G-CSF conjugates
ES2327606T3 (en) 2000-01-10 2009-11-02 Maxygen Holdings Ltd CONJUGATES OF G-CSF.
US7049761B2 (en) * 2000-02-11 2006-05-23 Altair Engineering, Inc. Light tube and power supply circuit
ES2325877T3 (en) 2000-02-11 2009-09-23 Bayer Healthcare Llc FACTOR TYPE MOLECULES VII OR VIIA.
ES2220673T3 (en) 2000-02-29 2004-12-16 Pfizer Products Inc. STIMULATING FACTOR OF STABILIZED GRANULOCIT COLONIES.
US20020064820A1 (en) * 2000-03-13 2002-05-30 Jean-Michel Dayer Apo-A-I regulation of T-cell signaling
WO2001076639A2 (en) * 2000-04-06 2001-10-18 Pharmacia Corporation Chemically-modified myelopoietin conjugates
US6586398B1 (en) * 2000-04-07 2003-07-01 Amgen, Inc. Chemically modified novel erythropoietin stimulating protein compositions and methods
US6939541B2 (en) * 2000-04-14 2005-09-06 University Of South Carolina Cloning, overexpression and therapeutic use of bioactive histidine ammonia lyase
PT1290013E (en) 2000-04-21 2006-06-30 Amgen Inc DERIVATIVES OF APO-AI / AII PEPTIDES
US6677136B2 (en) 2000-05-03 2004-01-13 Amgen Inc. Glucagon antagonists
US20030211094A1 (en) * 2001-06-26 2003-11-13 Nelsestuen Gary L. High molecular weight derivatives of vitamin k-dependent polypeptides
MXPA03000311A (en) * 2000-07-12 2004-12-13 Gryphon Therapeutics Inc Chemokine receptor modulators, production and use.
US7118737B2 (en) * 2000-09-08 2006-10-10 Amylin Pharmaceuticals, Inc. Polymer-modified synthetic proteins
US7030218B2 (en) * 2000-09-08 2006-04-18 Gryphon Therapeutics Pseudo native chemical ligation
WO2002026265A2 (en) * 2000-09-29 2002-04-04 Schering Corporation Pegylated interleukin-10
EP1333036B1 (en) * 2000-10-16 2009-01-21 Chugai Seiyaku Kabushiki Kaisha Peg-modified erythropoietin
ES2382636T3 (en) * 2000-10-31 2012-06-12 Surmodics Pharmaceuticals, Inc. Method for producing compositions for improved administration of bioactive molecules
AU2002212108A1 (en) * 2000-11-02 2002-05-15 Maxygen Aps Single-chain multimeric polypeptides
CN1321134C (en) * 2000-11-23 2007-06-13 赵剑 Hetergeneous product of bio-active protein and its preparing process
MXPA03005388A (en) * 2000-12-14 2003-09-25 Amylin Pharmaceuticals Inc Peptide yy and peptide yy agonists for treatment of metabolic disorders.
US7053150B2 (en) 2000-12-18 2006-05-30 Nektar Therapeutics Al, Corporation Segmented polymers and their conjugates
TW593427B (en) * 2000-12-18 2004-06-21 Nektar Therapeutics Al Corp Synthesis of high molecular weight non-peptidic polymer derivatives
CN100528235C (en) * 2000-12-20 2009-08-19 霍夫曼-拉罗奇有限公司 Conjugates of erythropoietin
US7442370B2 (en) 2001-02-01 2008-10-28 Biogen Idec Ma Inc. Polymer conjugates of mutated neublastin
MXPA03007002A (en) * 2001-02-06 2003-11-18 Merck Patent Gmbh Modified granulocyte colony stimulating factor (g-csf) with reduced immunogenicity.
EP1234583A1 (en) 2001-02-23 2002-08-28 F. Hoffmann-La Roche Ag PEG-conjugates of HGF-NK4
WO2002074806A2 (en) 2001-02-27 2002-09-26 Maxygen Aps New interferon beta-like molecules
US7276580B2 (en) 2001-03-12 2007-10-02 Biogen Idec Ma Inc. Neurotrophic factors
CA2445947A1 (en) * 2001-04-30 2002-11-07 Targeted Genetics Corporation Lipid-comprising drug delivery complexes and methods for their production
US7247618B2 (en) * 2001-04-30 2007-07-24 Tripathi Rajavashisth Methods for inhibiting macrophage colony stimulating factor and c-FMS-dependent cell signaling
PT1385882E (en) 2001-05-11 2008-01-11 Amgen Inc Peptides and related molecules that bind to tall-1
CA2450985A1 (en) * 2001-06-28 2003-01-09 Mountain View Pharmaceuticals, Inc. Polymer stabilized proteinases
JP4444652B2 (en) * 2001-07-11 2010-03-31 マキシゲン・ホールディングズ・リミテッド G-CSF conjugate
US20040077835A1 (en) * 2001-07-12 2004-04-22 Robin Offord Chemokine receptor modulators, production and use
CA2460690A1 (en) * 2001-10-05 2003-04-17 Intermune, Inc. Method of treating hepatitis virus infection with a multiphasic interferon delivery profile
US7084257B2 (en) 2001-10-05 2006-08-01 Amgen Inc. Fully human antibody Fab fragments with human interferon-gamma neutralizing activity
US6908963B2 (en) * 2001-10-09 2005-06-21 Nektar Therapeutics Al, Corporation Thioester polymer derivatives and method of modifying the N-terminus of a polypeptide therewith
US7214660B2 (en) * 2001-10-10 2007-05-08 Neose Technologies, Inc. Erythropoietin: remodeling and glycoconjugation of erythropoietin
US7173003B2 (en) 2001-10-10 2007-02-06 Neose Technologies, Inc. Granulocyte colony stimulating factor: remodeling and glycoconjugation of G-CSF
US7157277B2 (en) 2001-11-28 2007-01-02 Neose Technologies, Inc. Factor VIII remodeling and glycoconjugation of Factor VIII
US7138370B2 (en) 2001-10-11 2006-11-21 Amgen Inc. Specific binding agents of human angiopoietin-2
SI1436012T1 (en) 2001-10-18 2018-03-30 Nektar Therapeutics Polymer conjugates of opioid antagonists
JPWO2003043651A1 (en) * 2001-11-19 2005-03-10 協和醗酵工業株式会社 Drugs that mobilize multipotent stem cells from tissues to peripheral blood
PL374354A1 (en) * 2001-11-20 2005-10-17 Pharmacia Corporation Chemically-modified human growth hormone conjugates
US20030171285A1 (en) * 2001-11-20 2003-09-11 Finn Rory F. Chemically-modified human growth hormone conjugates
KR100480432B1 (en) * 2001-12-04 2005-04-06 선바이오(주) Conjugates of granulocyte-colony stimulating factor and polyethylene glycol derivatives
WO2003049760A1 (en) * 2001-12-07 2003-06-19 Intermune, Inc. Compositions and method for treating hepatitis virus infection
US7838619B2 (en) * 2002-01-14 2010-11-23 The General Hospital Corporation Biodegradable polyketal polymers and methods for their formation and use
MEP35608A (en) 2002-01-18 2011-02-10 Biogen Idec Inc Polyalkylene polymer compounds and uses thereof
DE10209821A1 (en) 2002-03-06 2003-09-25 Biotechnologie Ges Mittelhesse Coupling of proteins to a modified polysaccharide
US20030191056A1 (en) 2002-04-04 2003-10-09 Kenneth Walker Use of transthyretin peptide/protein fusions to increase the serum half-life of pharmacologically active peptides/proteins
US20030199464A1 (en) * 2002-04-23 2003-10-23 Silviu Itescu Regeneration of endogenous myocardial tissue by induction of neovascularization
AU2003243545A1 (en) * 2002-06-14 2003-12-31 Amylin Pharmaceuticals, Inc. Prevention and/or treatment of inflammatory bowel disease using pyy or agonists thereof
US20040002451A1 (en) * 2002-06-20 2004-01-01 Bruce Kerwin Compositions of pegylated soluble tumor necrosis factor receptors and methods of preparing
EP1534269B1 (en) 2002-07-19 2013-10-30 The General Hospital Corporation Oxime conjugates and methods for their formation and use
AU2003256613A1 (en) * 2002-07-19 2004-02-09 Amgen Inc. Protein conjugates with a water-soluble biocompatible, biogradable polymer
JP2006508056A (en) 2002-08-28 2006-03-09 イミュネックス・コーポレーション Compositions and methods for treating cardiovascular disease
KR100974843B1 (en) * 2002-09-09 2010-08-11 넥타르 테라퓨틱스 Water-soluble polymer alkanals
US8129330B2 (en) 2002-09-30 2012-03-06 Mountain View Pharmaceuticals, Inc. Polymer conjugates with decreased antigenicity, methods of preparation and uses thereof
JP2004196770A (en) * 2002-10-24 2004-07-15 Effector Cell Institute Inc Agent for increasing blood level of dendritic cell precursor
US20050075279A1 (en) * 2002-10-25 2005-04-07 Boehringer Ingelheim International Gmbh Macrocyclic peptides active against the hepatitis C virus
US7314613B2 (en) 2002-11-18 2008-01-01 Maxygen, Inc. Interferon-alpha polypeptides and conjugates
PA8588901A1 (en) * 2002-11-20 2005-02-04 Pharmacia Corp CONJUGATES OF N-TERMINAL HUMAN GROWTH HORMONE HORMONE AND PROCESS FOR PREPARATION
GEP20084486B (en) * 2002-12-26 2008-09-25 Mountain View Pharmaceuticals Polymer conjugates of interferon-beta with enhanced biological potency
AU2003303636B2 (en) * 2002-12-26 2010-08-05 Mountain View Pharmaceuticals, Inc. Polymer conjugates of cytokines, chemokines, growth factors, polypeptide hormones and antagonists thereof with preserved receptor-binding activity
EP1577323B1 (en) 2002-12-26 2013-07-10 Takeda Pharmaceutical Company Limited Metastin derivative and use thereof
EP1616003A4 (en) 2002-12-30 2007-06-20 Gryphon Therapeutics Inc Water-soluble thioester and selenoester compounds and methods for making and using the same
US7803362B2 (en) * 2003-01-24 2010-09-28 Synageva Biopharma Corp. Glycosylated interferon alpha
CA2516552A1 (en) * 2003-02-26 2004-09-10 Intermune, Inc. Polyethylene glycol modified interferon compositions and methods of use thereof
WO2004078127A2 (en) * 2003-02-28 2004-09-16 Intermune, Inc. Continuous delivery methods for treating hepatitis virus infection
US7871607B2 (en) 2003-03-05 2011-01-18 Halozyme, Inc. Soluble glycosaminoglycanases and methods of preparing and using soluble glycosaminoglycanases
BRPI0408116B1 (en) 2003-03-05 2022-09-20 Halozyme, Inc POLYMER-CONJUGATED SOLUBLE HYALURONIDASE POLYPEPTIDES, PHARMACEUTICAL COMPOSITIONS COMPRISING SOLUBLE PH20 POLYPEPTIDES, THEIR USES AND PREPARATION PROCESS, AND NUCLEIC ACIDS ENCODING SOLUBLE HYALURONIDASE POLYPEPTIDES
US20060104968A1 (en) 2003-03-05 2006-05-18 Halozyme, Inc. Soluble glycosaminoglycanases and methods of preparing and using soluble glycosaminogly ycanases
CA2519092C (en) 2003-03-14 2014-08-05 Neose Technologies, Inc. Branched water-soluble polymers and their conjugates
US20060134736A1 (en) * 2003-03-28 2006-06-22 Jacobs John W Human growth hormone conjugated with biocompatible polymer
MXPA05010411A (en) * 2003-03-28 2006-05-31 Biopolymed Inc Biologically active material conjugated with biocompatible polymer with 1:1 complex, preparation method thereof and pharmaceutical composition comprising the same.
US7587286B2 (en) 2003-03-31 2009-09-08 Xencor, Inc. Methods for rational pegylation of proteins
US7642340B2 (en) 2003-03-31 2010-01-05 Xencor, Inc. PEGylated TNF-α variant proteins
US7610156B2 (en) 2003-03-31 2009-10-27 Xencor, Inc. Methods for rational pegylation of proteins
US20070026485A1 (en) 2003-04-09 2007-02-01 Neose Technologies, Inc. Glycopegylation methods and proteins/peptides produced by the methods
US8791070B2 (en) 2003-04-09 2014-07-29 Novo Nordisk A/S Glycopegylated factor IX
WO2006127896A2 (en) 2005-05-25 2006-11-30 Neose Technologies, Inc. Glycopegylated factor ix
CA2521381C (en) 2003-04-11 2020-05-26 Kenneth Hinds Method for preparation of site-specific protein conjugates
EP2261244A3 (en) 2003-04-15 2011-02-23 Glaxosmithkline LLC Human il-18 substitution mutants and their conjugates
CA2864810A1 (en) 2003-04-18 2004-11-04 Biogen Idec Ma, Inc. Polymer-conjugated glycosylated neublastin
JP2007525188A (en) 2003-05-16 2007-09-06 インターミューン インコーポレイテッド Synthetic chemokine receptor ligands and methods of use thereof
CA2533702C (en) 2003-07-22 2012-05-22 Nektar Therapeutics Al, Corporation Method for preparing functionalized polymers from polymer alcohols
WO2005012484A2 (en) 2003-07-25 2005-02-10 Neose Technologies, Inc. Antibody-toxin conjugates
EP2133362B1 (en) 2003-07-25 2012-04-18 Amgen, Inc Methods relating to LDCAM and CRTAM
WO2005014655A2 (en) 2003-08-08 2005-02-17 Fresenius Kabi Deutschland Gmbh Conjugates of hydroxyalkyl starch and a protein
US7691975B2 (en) 2003-08-25 2010-04-06 Toray Industries, Inc. Interferon-β complex
GB0320638D0 (en) 2003-09-03 2003-10-01 Novartis Ag Organic compounds
CN1852740B (en) 2003-09-17 2011-05-11 耐科塔医药公司 Multi-arm polymer prodrugs
CN1856502A (en) * 2003-09-22 2006-11-01 贝林格尔.英格海姆国际有限公司 Macrocyclic peptides active against the hepatitis c virus
ES2737837T3 (en) * 2003-10-09 2020-01-16 Ambrx Inc Polymeric derivatives
WO2005035565A1 (en) 2003-10-10 2005-04-21 Novo Nordisk A/S Il-21 derivatives
WO2005035564A2 (en) 2003-10-10 2005-04-21 Xencor, Inc. Protein based tnf-alpha variants for the treatment of tnf-alpha related disorders
CA2540858C (en) 2003-10-14 2009-12-08 Intermune, Inc. Macrocyclic carboxylic acids and acylsulfonamides as inhibitors of hcv replication
EP2641611A3 (en) 2003-10-17 2013-12-18 Novo Nordisk A/S Combination therapy
WO2005040758A2 (en) * 2003-10-24 2005-05-06 Intermune, Inc. Use of pirfenidone in therapeutic regimens
US7220407B2 (en) * 2003-10-27 2007-05-22 Amgen Inc. G-CSF therapy as an adjunct to reperfusion therapy in the treatment of acute myocardial infarction
EP1725572B1 (en) 2003-11-05 2017-05-31 AGCT GmbH Macromolecular nucleotide compounds and methods for using the same
US20080305992A1 (en) 2003-11-24 2008-12-11 Neose Technologies, Inc. Glycopegylated erythropoietin
US8633157B2 (en) 2003-11-24 2014-01-21 Novo Nordisk A/S Glycopegylated erythropoietin
US20060040856A1 (en) 2003-12-03 2006-02-23 Neose Technologies, Inc. Glycopegylated factor IX
WO2005053730A1 (en) * 2003-12-05 2005-06-16 Kirin Beer Kabushiki Kaisha Therapeutic agent for terminal heart failure
WO2005067963A1 (en) * 2003-12-23 2005-07-28 Intermune, Inc. Use of polyethylene glycol-modified interferon-alpha in therapeutic dosing regimens
GB0500020D0 (en) 2005-01-04 2005-02-09 Novartis Ag Organic compounds
ES2560657T3 (en) 2004-01-08 2016-02-22 Ratiopharm Gmbh O-linked glycosylation of G-CSF peptides
EP1730167B1 (en) * 2004-01-21 2011-01-12 Boehringer Ingelheim International GmbH Macrocyclic peptides active against the hepatitis c virus
BRPI0507169A (en) 2004-02-02 2007-06-26 Ambrx Inc modified human growth hormone polypeptides and their uses
AU2005211776B2 (en) 2004-02-11 2012-02-02 Amylin Pharmaceuticals, Llc Pancreatic polypeptide family motifs and polypeptides comprising the same
US8076288B2 (en) 2004-02-11 2011-12-13 Amylin Pharmaceuticals, Inc. Hybrid polypeptides having glucose lowering activity
US20060094652A1 (en) 2004-02-11 2006-05-04 Levy Odile E Hybrid polypeptides with selectable properties
JP2007526317A (en) * 2004-03-01 2007-09-13 エンゾン ファーマスーティカルズ インコーポレイテッド Interferon-beta polymer conjugate
CA2560289A1 (en) * 2004-03-23 2005-10-13 Amgen Inc. Chemically modified protein compositions and methods
US8470315B2 (en) * 2004-04-13 2013-06-25 Quintessence Biosciences, Inc. Non-natural ribonuclease conjugates as cytotoxic agents
EP1586334A1 (en) * 2004-04-15 2005-10-19 TRASTEC scpa G-CSF conjugates with peg
BRPI0511196A (en) 2004-05-19 2007-12-04 Maxygen Inc isolated or recombinant polypeptide, conjugate, composition, host cell, vector, methods for making a polypeptide, for making a conjugate, for reducing the copy number of a virus in virus-infected cells, for reducing the level of rna hcv, dna hbv and rna hiv in the serum of a patient infected with hcv, hbv and hiv, and use of the polypeptide, conjugate or composition
WO2005123113A2 (en) * 2004-06-14 2005-12-29 Intermune, Inc. Interferon compositions and methods of use thereof
JP2008503217A (en) 2004-06-18 2008-02-07 アンブレツクス・インコーポレイテツド Novel antigen-binding polypeptides and their use
CN101845091A (en) 2004-06-25 2010-09-29 武田药品工业株式会社 Metastin derivatives and use thereof
JP2008509889A (en) * 2004-06-30 2008-04-03 イージェン コーポレーション PEGylated interferon alpha-1b
CA2572765C (en) * 2004-07-08 2013-05-21 Amgen Inc. Compound having improved bioefficiency when administered in a multidose regimen
WO2006010143A2 (en) * 2004-07-13 2006-01-26 Neose Technologies, Inc. Branched peg remodeling and glycosylation of glucagon-like peptide-1 [glp-1]
AU2005327906B2 (en) 2004-07-21 2010-05-13 Ambrx, Inc. Biosynthetic polypeptides utilizing non-naturally encoded amino acids
US7597884B2 (en) 2004-08-09 2009-10-06 Alios Biopharma, Inc. Hyperglycosylated polypeptide variants and methods of use
AU2005277227B2 (en) 2004-08-19 2011-10-06 Biogen Ma Inc. Refolding transforming growth factor beta family proteins
ATE474586T1 (en) 2004-08-19 2010-08-15 Biogen Idec Inc NEUBLASTIN VARIANTS
EP1799249A2 (en) 2004-09-10 2007-06-27 Neose Technologies, Inc. Glycopegylated interferon alpha
AU2005286662B2 (en) 2004-09-23 2011-10-06 Vasgene Therapeutics, Inc. Polypeptide compounds for inhibiting angiogenesis and tumor growth
DK2586456T3 (en) 2004-10-29 2016-03-21 Ratiopharm Gmbh Conversion and glycopegylation of fibroblast growth factor (FGF)
WO2006055260A2 (en) 2004-11-05 2006-05-26 Northwestern University Use of scf and g-csf in the treatment of cerebral ischemia and neurological disorders
BRPI0518559A2 (en) 2004-12-13 2008-11-25 Amylin Pharmaceuticals Inc motifs of the pancreatic polypeptide family, polypeptides and methods comprising the same
EP1674113A1 (en) 2004-12-22 2006-06-28 F. Hoffmann-La Roche Ag Conjugates of insulin-like growth factor-1 (IGF-1) and poly(ethylene glycol)
EP1836314B1 (en) 2004-12-22 2012-01-25 Ambrx, Inc. Modified human growth hormone
CA2594561C (en) 2004-12-22 2014-12-23 Ambrx, Inc. Formulations of human growth hormone comprising a non-naturally encoded amino acid
CA2590429C (en) * 2004-12-22 2014-10-07 Ambrx, Inc. Compositions of aminoacyl-trna synthetase and uses thereof
NZ555206A (en) 2004-12-22 2010-09-30 Ambrx Inc Methods for expression and purification of recombinant human growth hormone
JP5425398B2 (en) 2004-12-22 2014-02-26 アンブレツクス・インコーポレイテツド Compositions comprising unnatural amino acids and polypeptides, methods relating to unnatural amino acids and polypeptides, and uses of unnatural amino acids and polypeptides
JP4951527B2 (en) 2005-01-10 2012-06-13 バイオジェネリックス アーゲー GlycoPEGylated granulocyte colony stimulating factor
CA2595695A1 (en) 2005-01-25 2006-08-03 Cell Therapeutics, Inc. Biologically active protein conjugates comprising (nn[s/t]) peptide repeats and having increased plasma half-life
AU2005326226B2 (en) * 2005-01-31 2010-11-11 Eci, Inc. Immunopotentiating agent
US7402730B1 (en) 2005-02-03 2008-07-22 Lexicon Pharmaceuticals, Inc. Knockout animals manifesting hyperlipidemia
US8263545B2 (en) 2005-02-11 2012-09-11 Amylin Pharmaceuticals, Inc. GIP analog and hybrid polypeptides with selectable properties
EA011653B1 (en) * 2005-02-11 2009-04-28 Амилин Фармасьютикалз, Инк. Gip analog and hybrid polypeptides with selectable properties
WO2006094530A1 (en) * 2005-03-11 2006-09-14 Siegfried Ltd. Di-polymer protein conjugates and processes for their preparation
WO2007010552A2 (en) * 2005-03-17 2007-01-25 Serum Institute Of India Limited N- terminal peg conjugate of erythropoietin
EP1871403B1 (en) 2005-03-31 2010-09-08 Amylin Pharmaceuticals, Inc. Compositions and methods for the control, prevention and treatment of eating disorders
EP2314320A2 (en) 2005-04-05 2011-04-27 Istituto di Richerche di Biologia Molecolare P. Angeletti S.p.A. Method for shielding functional sites or epitopes on proteins
US20070154992A1 (en) 2005-04-08 2007-07-05 Neose Technologies, Inc. Compositions and methods for the preparation of protease resistant human growth hormone glycosylation mutants
LT3321359T (en) 2005-04-11 2021-05-10 Horizon Pharma Rheumatology Llc Variant forms of urate oxidase and use thereof
US7833979B2 (en) 2005-04-22 2010-11-16 Amgen Inc. Toxin peptide therapeutic agents
US7619067B2 (en) 2005-05-18 2009-11-17 Maxygen, Inc. Evolved interferon-alpha polypeptides
WO2006130799A2 (en) * 2005-06-01 2006-12-07 Alza Corporation Bioconjugation reactions for acylating polyethlene glycol reagents
WO2006128460A2 (en) * 2005-06-01 2006-12-07 Maxygen Holdings Ltd. Pegylated g-csf polypeptides and methods of producing same
WO2007009208A1 (en) * 2005-06-02 2007-01-25 Cangene Corporation Poly(ethylene glocol) modified human gm-csf with increased biological activity
CN103030690A (en) * 2005-06-03 2013-04-10 Ambrx公司 Improved human interferon molecules and their uses
AU2006257792A1 (en) * 2005-06-13 2006-12-21 Mdrna, Inc. Transmucosal delivery of peptide derivatives
KR100694994B1 (en) * 2005-06-13 2007-03-14 씨제이 주식회사 Human Granulocyte-Colony Stimulating Factor Isoforms
ATE524183T1 (en) 2005-06-17 2011-09-15 Novartis Ag USE OF SANGLIFEHRIN IN HCV THERAPY
EP1893632B1 (en) 2005-06-17 2015-08-12 Novo Nordisk Health Care AG Selective reduction and derivatization of engineered factor vii proteins comprising at least one non-native cysteine
US7695710B2 (en) * 2005-06-20 2010-04-13 Pepgen Corporation Antitumor and antiviral combination therapies using low-toxicity, long-circulating human interferon-alpha analogs
TW200722104A (en) * 2005-06-20 2007-06-16 Pepgen Corp Low-toxicity, long-circulating human interferon-α analogs
WO2007011802A1 (en) * 2005-07-18 2007-01-25 Nektar Therapeutics Al, Corporation Method for preparing branched functionalized polymers using branched polyol cores
KR100735784B1 (en) 2005-07-20 2007-07-06 재단법인 목암생명공학연구소 Mutant of granulocyte-colony stimulating factorG-CSF and chemically conjugated polypeptide thereof
GEP20105124B (en) 2005-07-25 2010-11-25 Array Biopharma Inc Novel macrocyclic inhibitors of hepatitis c virus replication
CN101208386B (en) * 2005-08-02 2012-07-18 第一毛织株式会社 Epoxy resin composition for sealing semiconductor device
AU2006278490A1 (en) * 2005-08-04 2007-02-15 Nektar Therapeutics Conjugates of a G-CSF moiety and a polymer
BRPI0614649A2 (en) 2005-08-11 2011-04-12 Amylin Pharmaceuticals Inc hybrid polypeptides with selectable properties
EP1922336B1 (en) * 2005-08-11 2012-11-21 Amylin Pharmaceuticals, LLC Hybrid polypeptides with selectable properties
US8008453B2 (en) 2005-08-12 2011-08-30 Amgen Inc. Modified Fc molecules
ES2408581T3 (en) 2005-08-18 2013-06-21 Ambrx, Inc. TRNA compositions and uses thereof
US20070105755A1 (en) 2005-10-26 2007-05-10 Neose Technologies, Inc. One pot desialylation and glycopegylation of therapeutic peptides
MX2008002370A (en) 2005-08-19 2008-04-29 Amylin Pharmaceuticals Inc Exendin for treating diabetes and reducing body weight.
KR20080056295A (en) 2005-10-11 2008-06-20 인터뮨, 인크. Compounds and methods for inhibiting hepatitis c viral replication
WO2007047303A2 (en) * 2005-10-12 2007-04-26 Alvine Pharmaceuticals, Inc. Pegylated glutenase polypeptides
EP3037544A1 (en) 2005-10-13 2016-06-29 Human Genome Sciences, Inc. Methods and compositions for use in treatment of systemic lupus erythematosus (sle) patients with autoantibody positive diseases
US20070092486A1 (en) * 2005-10-21 2007-04-26 Avigenics, Inc. Glycolated and glycosylated poultry derived therapeutic proteins
US8168592B2 (en) 2005-10-21 2012-05-01 Amgen Inc. CGRP peptide antagonists and conjugates
US20080171696A1 (en) * 2005-10-21 2008-07-17 Avigenics, Inc. Pharmacodynamically enhanced therapeutic proteins
US20090048440A1 (en) 2005-11-03 2009-02-19 Neose Technologies, Inc. Nucleotide Sugar Purification Using Membranes
JP5508716B2 (en) * 2005-11-08 2014-06-04 アンブルックス,インコーポレイテッド Accelerators for modifying unnatural amino acids and unnatural amino acid polypeptides
PE20070684A1 (en) 2005-11-14 2007-08-06 Amgen Inc RANKL-PTH / PTHrP ANTIBODY CHEMERICAL MOLECULES
US20090018029A1 (en) * 2005-11-16 2009-01-15 Ambrx, Inc. Methods and Compositions Comprising Non-Natural Amino Acids
CA2882445A1 (en) * 2005-12-14 2007-06-21 Ambrx, Inc. Compositions containing, methods involving, and uses of non-natural amino acids and polypeptides
US8404643B2 (en) 2005-12-22 2013-03-26 Takeda Pharmaceutical Company Limited Metastin derivatives and use thereof
ES2664086T3 (en) 2005-12-30 2018-04-18 Zensun (Shanghai) Science & Technology, Co., Ltd. Extended release of neurregulin to improve cardiac function
US8992905B2 (en) 2006-01-12 2015-03-31 Hokusan Co. Ltd. Oral composition containing interferon-α
CN100475270C (en) * 2006-01-20 2009-04-08 清华大学 Medicine for treating tumor, and application thereof
WO2007088051A2 (en) 2006-01-31 2007-08-09 Bayer Schering Pharma Aktiengesellschaft Modulation of mdl-1 activity for treatment of inflammatory disease
EP2319542B1 (en) 2006-02-21 2018-03-21 Nektar Therapeutics Segmented degradable polymers and conjugates made therefrom
TWI501774B (en) 2006-02-27 2015-10-01 Biogen Idec Inc Treatments for neurological disorders
JP2009529915A (en) 2006-03-20 2009-08-27 ゾーマ テクノロジー リミテッド Human antibodies and methods specific for gastrin substances
CA2648053A1 (en) * 2006-03-31 2007-10-11 Centocor, Inc. Binding partners with immunoglobulin domains modified to have extended half-life
EP2007789B1 (en) 2006-04-11 2015-05-20 Novartis AG Spirocyclic HCV/HIV inhibitors and their uses
US9283260B2 (en) * 2006-04-21 2016-03-15 Amgen Inc. Lyophilized therapeutic peptibody formulations
US8299024B2 (en) * 2006-05-12 2012-10-30 Amylin Pharmaceuticals, Llc Methods to restore glycemic control
US7851438B2 (en) 2006-05-19 2010-12-14 GlycoFi, Incorporated Erythropoietin compositions
RU2008145084A (en) 2006-05-24 2010-06-27 Ново Нордиск Хелс Кеа Аг (Ch) ANALOGUES OF FACTOR IX HAVING A LONG-TERM HALF-TIME IN VIVO
US7981425B2 (en) 2006-06-19 2011-07-19 Amgen Inc. Thrombopoietic compounds
EP2044097A4 (en) * 2006-06-23 2010-10-06 Quintessence Biosciences Inc Modified ribonucleases
CA2787343C (en) 2006-06-26 2016-08-02 Amgen Inc. Compositions comprising modified lcat and uses thereof
WO2008010991A2 (en) * 2006-07-17 2008-01-24 Quintessence Biosciences, Inc. Methods and compositions for the treatment of cancer
WO2008011633A2 (en) 2006-07-21 2008-01-24 Neose Technologies, Inc. Glycosylation of peptides via o-linked glycosylation sequences
JP5419689B2 (en) * 2006-07-25 2014-02-19 リポクセン テクノロジーズ リミテッド Derivatization of granulocyte colony-stimulating factor
GB0615067D0 (en) * 2006-07-28 2006-09-06 Ttp Communications Ltd Reconfigurable signal processing scheme
ES2529234T3 (en) * 2006-08-04 2015-02-18 Prolong Pharmaceuticals, LLC Modified erythropoietin
ITMI20061624A1 (en) * 2006-08-11 2008-02-12 Bioker Srl SINGLE-CONJUGATE SITE-SPECIFIC OF G-CSF
US8497240B2 (en) 2006-08-17 2013-07-30 Amylin Pharmaceuticals, Llc DPP-IV resistant GIP hybrid polypeptides with selectable properties
CL2007002502A1 (en) 2006-08-31 2008-05-30 Hoffmann La Roche VARIANTS OF THE SIMILAR GROWTH FACTOR TO HUMAN INSULIN-1 (IGF-1) PEGILATED IN LISIN; METHOD OF PRODUCTION; FUSION PROTEIN THAT UNDERSTANDS IT; AND ITS USE TO TREAT ALZHEIMER'S DISEASE.
MX2009001691A (en) * 2006-08-31 2009-02-25 Hoffmann La Roche Method for the production of insulin-like growth factor-i.
WO2008030614A2 (en) * 2006-09-08 2008-03-13 Ambrx, Inc. Suppressor trna transcription in vertebrate cells
AU2007292892B9 (en) * 2006-09-08 2013-02-28 Ambrx, Inc. Hybrid suppressor tRNA for vertebrate cells
JP5840345B2 (en) 2006-09-08 2016-01-06 アンブルックス, インコーポレイテッドAmbrx, Inc. Modified human plasma polypeptides or modified human Fc scaffold proteins and uses thereof
US7985783B2 (en) 2006-09-21 2011-07-26 The Regents Of The University Of California Aldehyde tags, uses thereof in site-specific protein modification
US20080081031A1 (en) 2006-09-28 2008-04-03 Schering Corporation Use of Pegylated IL-10 to Treat Cancer
US8969532B2 (en) 2006-10-03 2015-03-03 Novo Nordisk A/S Methods for the purification of polypeptide conjugates comprising polyalkylene oxide using hydrophobic interaction chromatography
SI2068907T1 (en) * 2006-10-04 2018-01-31 Novo Nordisk A/S Glycerol linked pegylated sugars and glycopeptides
WO2008051383A2 (en) * 2006-10-19 2008-05-02 Amgen Inc. Use of alcohol co-solvents to improve pegylation reaction yields
JO3048B1 (en) 2006-10-25 2016-09-05 Takeda Pharmaceuticals Co Metastin Derivatives And Use Thereof
WO2008088422A2 (en) 2006-10-25 2008-07-24 Amgen Inc. Toxin peptide therapeutic agents
KR101079993B1 (en) * 2006-11-17 2011-11-04 동아제약주식회사 Polyethylene glycol-G-CSF conjugate
EP3156415A1 (en) 2006-11-22 2017-04-19 Bristol-Myers Squibb Company Targeted therapeutics based on engineered proteins for tyrosine kinases receptors, including igf-ir
ES2618830T3 (en) 2006-12-08 2017-06-22 Lexicon Pharmaceuticals, Inc. Monoclonal antibodies against ANGPTL3
EP2111228B1 (en) 2007-02-02 2011-07-20 Bristol-Myers Squibb Company 10Fn3 domain for use in treating diseases associated with inappropriate angiogenesis
CN101245109B (en) * 2007-02-12 2011-12-14 杭州九源基因工程有限公司 Polyglycol single-modificatory recombined human granular leukocyte colony stimulating factor mutant and preparation method thereof
KR101476472B1 (en) 2007-03-30 2015-01-05 암브룩스, 인코포레이티드 Modified fgf-21 polypeptides and their uses
CA2682897C (en) 2007-04-03 2016-11-22 Biogenerix Ag Methods of treatment using glycopegylated g-csf
JP2008266219A (en) * 2007-04-20 2008-11-06 National Institute Of Advanced Industrial & Technology Protein not containing lysine and cysteine residues
JP5583005B2 (en) 2007-05-01 2014-09-03 バイオジェン・アイデック・エムエイ・インコーポレイテッド Compositions and methods for increasing angiogenesis
JP2010525821A (en) 2007-05-02 2010-07-29 アンブルックス,インコーポレイテッド Modified IFN beta polypeptides and their use
US8420779B2 (en) 2007-05-22 2013-04-16 Amgen Inc. Compositions and methods for producing bioactive fusion proteins
CN101778937A (en) * 2007-06-04 2010-07-14 诺和诺德公司 o-linked glycosylation using n-acetylglucosaminyl transferases
MX2009013259A (en) 2007-06-12 2010-01-25 Novo Nordisk As Improved process for the production of nucleotide sugars.
WO2009003188A2 (en) * 2007-06-27 2008-12-31 Cedars-Sinai Medical Center N-terminal specific chemical labeling for proteomics applications
PT2181190E (en) 2007-07-26 2014-02-24 Amgen Inc Modified lecithin-cholesterol acyltransferase enzymes
CN101352573B (en) * 2007-07-27 2011-02-09 杭州九源基因工程有限公司 Recombinant human granulocyte colony stimulating factor lysine defect body modified by polyethyleneglycol
AU2008287063B2 (en) 2007-08-09 2013-10-24 Genzyme Corporation Method of treating autoimmune disease with mesenchymal stem cells
CL2008002399A1 (en) * 2007-08-16 2009-01-02 Pharmaessentia Corp Substantially pure conjugate having a polymeric portion, a protein portion (interferon alpha 2b) and an aliphatic binder of 1 to 10 carbon atoms, useful in the treatment of hepatitis b or c.
AU2008292186B9 (en) 2007-08-27 2014-07-03 Ratiopharm Gmbh Liquid formulation of G-CSF conjugate
US8207112B2 (en) 2007-08-29 2012-06-26 Biogenerix Ag Liquid formulation of G-CSF conjugate
US8758761B2 (en) * 2007-09-30 2014-06-24 University Of Florida Research Foundation, Inc. Combination therapies for treating type 1 diabetes
WO2009048909A1 (en) * 2007-10-08 2009-04-16 Quintessence Biosciences, Inc. Compositions and methods for ribonuclease-based therapies
MX2010004400A (en) 2007-10-23 2010-05-20 Nektar Therapeutics Hydroxyapatite-targeting multiarm polymers and conjugates made therefrom.
DK2217265T3 (en) 2007-11-20 2017-07-03 Ambrx Inc Modified insulin polypeptides and their use
WO2009073977A1 (en) * 2007-12-13 2009-06-18 Biovectra Inc. Polypeptides modified by protein trans-splicing technology
BRPI0821700A2 (en) * 2007-12-27 2015-06-16 Baxter Int Methods for determining the number of physiologically acceptable polymer molecules bound to a protein or protein complex in a polymer-protein conjugate and to determining the number of physiologically acceptable polymer molecules bound to a protein or protein complex or free in solution
US20100316702A1 (en) * 2008-01-08 2010-12-16 The Regents Of The University Of California Compositions and methods for regulating erythropoeitin expression and ameliorating anemia and stimulating erythropoiesis
US20100286067A1 (en) * 2008-01-08 2010-11-11 Biogenerix Ag Glycoconjugation of polypeptides using oligosaccharyltransferases
US20090183503A1 (en) * 2008-01-18 2009-07-23 Alberto Verdesi Exhaust apparatus
EP3219795B1 (en) 2008-01-18 2024-03-13 BioMarin Pharmaceutical Inc. Manufacture of active highly phosphorylated human lysosomal sulfatase enzymes and uses thereof
CN103694337B (en) 2008-02-08 2016-03-02 Ambrx公司 Modified leptin polypeptide and its purposes
US8629104B2 (en) 2008-02-18 2014-01-14 Jiangsu Hengrui Medicine Co. Ltd. G-CSF and water-soluble polymer conjugate
PL2257311T3 (en) 2008-02-27 2014-09-30 Novo Nordisk As Conjugated factor viii molecules
TWI395593B (en) 2008-03-06 2013-05-11 Halozyme Inc In vivo temporal control of activatable matrix-degrading enzymes
US9840546B2 (en) * 2008-04-03 2017-12-12 Biosteed Gene Expression Tech. Co., Ltd. Double-stranded polyethylene glycol modified growth hormone, preparation method and application thereof
AU2009231186A1 (en) * 2008-04-03 2009-10-08 F. Hoffmann-La Roche Ag Pegylated insulin-like-growth-factor assay
WO2009128917A2 (en) 2008-04-14 2009-10-22 Halozyme, Inc. Modified hyaluronidases and uses in treating hyaluronan-associated diseases and conditions
TWI394580B (en) 2008-04-28 2013-05-01 Halozyme Inc Super fast-acting insulin compositions
EP2303313B1 (en) 2008-05-21 2015-10-28 Amylin Pharmaceuticals, LLC Exendins to lower cholestrol and triglycerides
JP2011520961A (en) 2008-05-22 2011-07-21 ブリストル−マイヤーズ スクイブ カンパニー Scaffold domain protein based on multivalent fibronectin
JOP20190083A1 (en) 2008-06-04 2017-06-16 Amgen Inc Fgf21 mutant fusion polypeptides and uses thereof
GB0811743D0 (en) 2008-06-26 2008-07-30 Hemosol Biopharma Inc Composition
EP2671891A3 (en) 2008-06-27 2014-03-05 Amgen Inc. Ang-2 inhibition to treat multiple sclerosis
EP3225248B1 (en) * 2008-07-23 2023-06-07 Ambrx, Inc. Modified bovine g-csf polypeptides and their uses
US20110212890A1 (en) * 2008-07-30 2011-09-01 Takeda Pharmaceutical Company Limited Metastin derivative and use thereof
CN102131844B (en) * 2008-07-31 2016-03-02 药华医药股份有限公司 Peptide-polymer conjugates
CN102159250B (en) 2008-08-11 2014-08-06 尼克塔治疗公司 Multi-arm polymeric alkanoate conjugates
EP2331139B1 (en) 2008-09-11 2019-04-17 Nektar Therapeutics Polymeric alpha-hydroxy aldehyde and ketone reagents and conjugation method
KR101732054B1 (en) * 2008-09-26 2017-05-02 암브룩스, 인코포레이티드 Non-natural amino acid replication-dependent microorganisms and vaccines
AU2009296397B2 (en) 2008-09-26 2012-11-08 Ambrx Inc. Modified animal erythropoietin polypeptides and their uses
JP2012504423A (en) 2008-10-01 2012-02-23 クインテッセンス バイオサイエンシズ,インコーポレーテッド Therapeutic ribonuclease
EA032727B1 (en) 2008-10-10 2019-07-31 Амген Инк. Fgf21 mutant proteolysis-resistant polypeptide and use thereof
DK2349341T3 (en) 2008-10-15 2014-01-06 Baxter Int Pegylation of recombinant blood coagulation factors in the presence of bound antibodies
CA2738679A1 (en) 2008-10-17 2010-04-22 Baxter International Inc. Modified blood factors comprising a low degree of water soluble polymer
EP2337794B1 (en) * 2008-10-20 2013-07-31 USV Limited An improved process for pegylation of proteins
WO2010048184A2 (en) * 2008-10-21 2010-04-29 Baxter International Inc. Methods for determining active ingredients in pro-drug peg protein conjugates with releasable peg reagents (in vitro de-pegylation)
WO2010051335A1 (en) * 2008-10-31 2010-05-06 Amgen Inc. Materials and methods relating to stem cell mobilization by multi-pegylated granulocyte colony stimulating factor
IT1392655B1 (en) 2008-11-20 2012-03-16 Bio Ker S R L SITE-SPECIFIC MONOCONJUGATED INSULINOTROPIC GLP-1 PEPTIDES.
TWI496582B (en) 2008-11-24 2015-08-21 必治妥美雅史谷比公司 Bispecific egfr/igfir binding molecules
US8927249B2 (en) 2008-12-09 2015-01-06 Halozyme, Inc. Extended soluble PH20 polypeptides and uses thereof
KR20110104943A (en) 2008-12-11 2011-09-23 백스터 인터내셔널 인코포레이티드 Detection of physiologically acceptable polymer molecules using near infrared spectroscopy
EP2379115B1 (en) 2008-12-17 2017-10-25 Merck Sharp & Dohme Corp. Mono- and di-peg il-10 production; and uses
US8512690B2 (en) 2009-04-10 2013-08-20 Novartis Ag Derivatised proline containing peptide compounds as protease inhibitors
US20110182850A1 (en) 2009-04-10 2011-07-28 Trixi Brandl Organic compounds and their uses
JP2012525847A (en) 2009-05-05 2012-10-25 アムジエン・インコーポレーテツド FGF21 variants and uses thereof
PE20120358A1 (en) 2009-05-05 2012-04-26 Amgen Inc FGF21 MUTANTS AND USES OF THEM
AR078044A1 (en) 2009-05-20 2011-10-12 Biomarin Pharm Inc VARIANTS OF NATURAL PEPTIDES OF TYPE C
CA2764835A1 (en) 2009-06-17 2010-12-23 Amgen Inc. Chimeric fgf19 polypeptides and uses thereof
MX2011014040A (en) 2009-06-19 2012-06-01 Medimmune Llc Protease variants.
WO2010151823A1 (en) 2009-06-25 2010-12-29 Savient Pharmaceuticals Inc. Methods and kits for predicting infusion reaction risk and antibody-mediated loss of response by monitoring serum uric acid during pegylated uricase therapy
WO2011014882A1 (en) 2009-07-31 2011-02-03 Medtronic, Inc. CONTINUOUS SUBCUTANEOUS ADMINISTRATION OF INTERFERON-α TO HEPATITIS C INFECTED PATIENTS
JP5734985B2 (en) 2009-09-17 2015-06-17 バクスター・ヘルスケヤー・ソシエテ・アノニムBaxter Healthcare SA Stable co-formulations of hyaluronidase and immunoglobulin and methods for their use
EP2480578A4 (en) * 2009-09-25 2013-04-17 Vybion Inc Polypeptide modification
US8926976B2 (en) 2009-09-25 2015-01-06 Xoma Technology Ltd. Modulators
WO2011038301A2 (en) 2009-09-25 2011-03-31 Xoma Technology Ltd. Screening methods
US9315860B2 (en) 2009-10-26 2016-04-19 Genovoxx Gmbh Conjugates of nucleotides and method for the application thereof
NZ598465A (en) 2009-10-30 2013-10-25 Boehringer Ingelheim Int Dosage regimens for hcv combination therapy comprising bi201335, interferon alpha and ribavirin
CN102695723A (en) 2009-10-30 2012-09-26 中枢神经系统治疗学公司 Improved neurturin molecules
EP2504021A4 (en) 2009-11-23 2013-05-15 Amylin Pharmaceuticals Llc Polypeptide conjugate
CA2781682A1 (en) 2009-12-04 2011-06-09 Genentech, Inc. Multispecific antibodies, antibody analogs, compositions, and methods
UA109888C2 (en) 2009-12-07 2015-10-26 ANTIBODY OR ANTIBODILITY ANTIBODY OR ITS BINDING TO THE β-CLOTE, FGF RECEPTORS AND THEIR COMPLEXES
EP2805964A1 (en) 2009-12-21 2014-11-26 Ambrx, Inc. Modified bovine somatotropin polypeptides and their uses
NZ600363A (en) 2009-12-21 2014-07-25 Ambrx Inc Modified porcine somatotropin polypeptides and their uses
US20110152188A1 (en) * 2009-12-23 2011-06-23 Hanns-Christian Mahler Pharmaceutical compositions of igf/i proteins
CN102770449B (en) 2010-02-16 2016-02-24 诺沃—诺迪斯克有限公司 The Factor VlII molecule that the VWF with reduction combines
CA2791361C (en) * 2010-03-04 2018-06-12 Pfenex Inc. Method for producing soluble recombinant interferon protein without denaturing
EP2552949B1 (en) 2010-04-01 2016-08-17 Pfenex Inc. Methods for g-csf production in a pseudomonas host cell
US9517264B2 (en) 2010-04-15 2016-12-13 Amgen Inc. Human FGF receptor and β-Klotho binding proteins
JP6066900B2 (en) 2010-04-26 2017-01-25 エータイアー ファーマ, インコーポレイテッド Innovative discovery of therapeutic, diagnostic and antibody compositions related to protein fragments of cysteinyl tRNA synthetase
AU2011248614B2 (en) 2010-04-27 2017-02-16 Pangu Biopharma Limited Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of isoleucyl tRNA synthetases
CA2797271C (en) 2010-04-28 2021-05-25 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of alanyl trna synthetases
EP2563383B1 (en) 2010-04-29 2017-03-01 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of valyl trna synthetases
WO2011139854A2 (en) 2010-04-29 2011-11-10 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of asparaginyl trna synthetases
CN102234310B (en) * 2010-04-30 2017-02-08 杭州九源基因工程有限公司 Polyethylene glycol modified protein separating and purifying method
CA2797277C (en) 2010-05-03 2021-02-23 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of arginyl-trna synthetases
US9034321B2 (en) 2010-05-03 2015-05-19 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of phenylalanyl-alpha-tRNA synthetases
CN103140233B (en) 2010-05-03 2017-04-05 Atyr 医药公司 Treatment, diagnosis and the discovery of antibody compositions related to the protein fragments of methionyl-tRNA synthetase
CN102985103A (en) 2010-05-04 2013-03-20 Atyr医药公司 Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of p38 multi-tRNA synthetase complex
US20120135912A1 (en) 2010-05-10 2012-05-31 Perseid Therapeutics Llc Polypeptide inhibitors of vla4
JP6396656B2 (en) 2010-05-14 2018-09-26 エータイアー ファーマ, インコーポレイテッド Innovative discovery of therapeutic, diagnostic and antibody compositions related to protein fragments of phenylalanyl βtRNA synthetase
JP2013533217A (en) 2010-05-17 2013-08-22 セビックス・インコーポレイテッド PEGylated C-peptide
US9034598B2 (en) 2010-05-17 2015-05-19 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of leucyl-tRNA synthetases
EP2576615B1 (en) 2010-05-26 2016-03-30 Bristol-Myers Squibb Company Fibronectin based scaffold proteins having improved stability
CN103096913B (en) 2010-05-27 2017-07-18 Atyr 医药公司 Treatment, diagnosis and the innovation of antibody compositions related to the protein fragments of glutaminyl tRNA synzyme is found
AU2011261486B2 (en) 2010-06-01 2017-02-23 Pangu Biopharma Limited Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of lysyl-tRNA synthetases
US20120123338A1 (en) 2010-06-16 2012-05-17 Medtronic, Inc. Damping systems for stabilizing medications in drug delivery devices
JP2013529627A (en) 2010-06-24 2013-07-22 パンメド リミテッド Treatment of hepatitis C virus-related disease using hydroxychloroquine or a combination of hydroxychloroquine and an antiviral agent
JP6116479B2 (en) 2010-07-12 2017-04-19 エータイアー ファーマ, インコーポレイテッド Innovative discovery of therapeutic, diagnostic and antibody compositions related to protein fragments of glycyl-tRNA synthetase
JP2013532176A (en) 2010-07-15 2013-08-15 ノヴォ ノルディスク アー/エス Stabilized factor VIII variant
ES2661089T3 (en) 2010-07-20 2018-03-27 Halozyme Inc. Methods of treatment or prevention of adverse side effects associated with the administration of an anti-hyaluronan agent
AR082319A1 (en) 2010-07-22 2012-11-28 Biomarin Pharm Inc PRODUCTION OF A HIGHLY ACTIVE PHOSPHORILED HUMAN N-ACETILGALACTOSAMINE-6-SULPHATASE AND ITS USES
CA2807552A1 (en) 2010-08-06 2012-02-09 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
RU2446173C1 (en) * 2010-08-13 2012-03-27 Зао "Биокад" New functional, high-purity stable conjugate of granulocyte colony-stimulating factor (g-csf) and polyethylene glycol with prolonged biological action, applicable for medical purposes, and based immunobiological agent
DK2605789T3 (en) 2010-08-17 2019-09-16 Ambrx Inc MODIFIED RELAXIN POLYPEPTIDES AND APPLICATIONS THEREOF
US9567386B2 (en) 2010-08-17 2017-02-14 Ambrx, Inc. Therapeutic uses of modified relaxin polypeptides
WO2012027611A2 (en) 2010-08-25 2012-03-01 Atyr Pharma, Inc. INNOVATIVE DISCOVERY OF THERAPEUTIC, DIAGNOSTIC, AND ANTIBODY COMPOSITIONS RELATED TO PROTEIN FRAGMENTS OF TYROSYL-tRNA SYNTHETASES
EP3466968A1 (en) 2010-09-15 2019-04-10 Stichting Sanquin Bloedvoorziening Factor viii variants having a decreased cellular uptake
TWI480288B (en) 2010-09-23 2015-04-11 Lilly Co Eli Formulations for bovine granulocyte colony stimulating factor and variants thereof
ES2630031T3 (en) 2010-09-28 2017-08-17 Aegerion Pharmaceuticals, Inc. A chimeric pinniped-human leptin polypeptide with increased solubility
WO2012045704A1 (en) 2010-10-05 2012-04-12 Novartis Ag New treatments of hepatitis c virus infection
JP5909495B2 (en) 2010-10-08 2016-04-26 ノバルティス アーゲー Vitamin E formulation of sulfamide NS3 inhibitor
CN102453089B (en) * 2010-10-25 2014-06-04 北京凯因科技股份有限公司 Preparation and application of recombinant consensus interferon mutant polyethylene glycol conjugate
CN103933577B (en) * 2010-10-25 2014-12-10 北京凯因科技股份有限公司 Preparation and application of recombinant interferon variant polyethylene glycol conjugate
US9023791B2 (en) 2010-11-19 2015-05-05 Novartis Ag Fibroblast growth factor 21 mutations
EP2643019B1 (en) 2010-11-24 2019-01-02 Lexicon Pharmaceuticals, Inc. Antibodies to notum pectinacetylesterase
EP2646038A1 (en) 2010-11-30 2013-10-09 Novartis AG New treatments of hepatitis c virus infection
CN102485742A (en) * 2010-12-02 2012-06-06 山东新时代药业有限公司 Preparation method and separation and purification method of polyethylene glycol single modified recombinant human granulocyte-colony stimulating factor
WO2012083197A1 (en) 2010-12-17 2012-06-21 Nektar Therapeutics Water-soluble polymer conjugates of topotecan
EP2654795B1 (en) 2010-12-21 2018-03-07 Nektar Therapeutics Multi-arm polymeric prodrug conjugates of pemetrexed-based compounds
WO2012088445A1 (en) 2010-12-22 2012-06-28 Nektar Therapeutics Multi-arm polymeric prodrug conjugates of cabazitaxel-based compounds
US20130331443A1 (en) 2010-12-22 2013-12-12 Nektar Therapeutics Multi-arm polymeric prodrug conjugates of taxane-based compounds
WO2012109387A1 (en) 2011-02-08 2012-08-16 Halozyme, Inc. Composition and lipid formulation of a hyaluronan-degrading enzyme and the use thereof for treatment of benign prostatic hyperplasia
PE20140593A1 (en) 2011-03-16 2014-05-10 Amgen Inc POWERFUL AND SELECTIVE INHIBITORS OF NAV1.3 AND NAV1.7
JP2014509646A (en) * 2011-03-25 2014-04-21 ザ・トラスティーズ・オブ・コランビア・ユニバーシティー・イン・ザ・シティー・オブ・ニューヨーク PEGylated human HDL particles and method for producing the same
KR20140007927A (en) 2011-03-31 2014-01-20 노파르티스 아게 Alisporivir to treat hepatitis c virus infection
BR112013024809A2 (en) 2011-04-01 2016-09-06 Novartis Ag treatment for hepatitis B virus infection alone or in combination with hepatitis delta virus and associated liver diseases
RU2013150344A (en) 2011-04-13 2015-05-20 Новартис Аг TREATMENT OF HEPATITIS VIRUS INFECTION ALISPOVIR
US20140187488A1 (en) 2011-05-17 2014-07-03 Bristol-Myers Squibb Company Methods for maintaining pegylation of polypeptides
ES2894398T3 (en) 2011-06-03 2022-02-14 Xoma Technology Ltd Specific antibodies to TGF-beta
CA2839512C (en) 2011-06-17 2018-01-02 Halozyme, Inc. Continuous subcutaneous insulin infusion methods with a hyaluronan-degrading enzyme
US9993529B2 (en) 2011-06-17 2018-06-12 Halozyme, Inc. Stable formulations of a hyaluronan-degrading enzyme
WO2013003593A1 (en) 2011-06-28 2013-01-03 Alternative Innovative Technologies Llc Methods of use of hsp70 for increased performance or hsp70 related disorders
AR087020A1 (en) 2011-07-01 2014-02-05 Bayer Ip Gmbh RELAXIN FUSION POLIPEPTIDES AND USES OF THE SAME
CN103747807B (en) 2011-07-05 2016-12-07 比奥阿赛斯技术有限公司 P97 antibody conjugates and using method
PT2729160T (en) 2011-07-08 2019-07-08 Aegerion Pharmaceuticals Inc Engineered polypeptides having enhanced duration of action and reduced immunogenicity
CN102952067A (en) * 2011-08-30 2013-03-06 苏州欣诺科生物科技有限公司 Pyridoxal derivative for pegylation modification of N terminal of protein and preparation method and application thereof
AU2012301769B2 (en) 2011-08-31 2016-05-19 Amgen Inc. FGF21 for use in treating type 1 diabetes
WO2013040501A1 (en) 2011-09-16 2013-03-21 Pharmathene, Inc. Compositions and combinations of organophosphorus bioscavengers and hyaluronan-degrading enzymes, and uses thereof
JO3476B1 (en) 2011-09-26 2020-07-05 Novartis Ag Fusion proteins for treating metabolic disorders
US9458214B2 (en) 2011-09-26 2016-10-04 Novartis Ag Dual function fibroblast growth factor 21 proteins
US20140228281A1 (en) 2011-09-27 2014-08-14 Novartis Ag Alisporivr for treatment of hepatitis c virus infection
RU2014114849A (en) 2011-10-14 2015-11-20 Алтернатив Инновейтив Текнолоджиз Ллц Degradation-resistant derivatives of proteins of heat shock-70 (btsh70) and methods of their application (options)
JP6162707B2 (en) 2011-10-24 2017-07-12 ハロザイム インコーポレイテッド Companion diagnostics for antihyaluronan treatment and methods of use thereof
US10350139B2 (en) 2011-10-25 2019-07-16 Corning Incorporated Pharmaceutical glass packaging assuring pharmaceutical sterility
WO2013063258A1 (en) 2011-10-25 2013-05-02 Corning Incorporated Delamination resistant pharmaceutical glass containers containing active pharmaceutical ingredients
CN104053670A (en) 2011-10-31 2014-09-17 百时美施贵宝公司 Fibronectin binding domains with reduced immunogenicity
CA2855770A1 (en) 2011-11-17 2013-05-23 Cebix Ab Pegylated c-peptide
WO2013101509A2 (en) 2011-12-15 2013-07-04 Alternative Innovative Technologies Llc Hsp70 fusion protein conjugates and uses thereof
EP3130347B1 (en) 2011-12-30 2019-09-18 Halozyme, Inc. Ph20 polypeptide variants, formulations and uses thereof
CA2861402C (en) 2012-01-30 2021-10-12 Guy Casy Stabilized aqueous antibody compositions
WO2013123432A2 (en) 2012-02-16 2013-08-22 Atyr Pharma, Inc. Histidyl-trna synthetases for treating autoimmune and inflammatory diseases
US10172953B2 (en) 2012-02-27 2019-01-08 Amunix Operating Inc. XTEN conjugate compositions and methods of making same
US20150030564A1 (en) 2012-02-29 2015-01-29 Toray Industries, Inc. Inhibitory agent for body cavity fluid accumulation
DK2822575T3 (en) 2012-03-03 2020-06-15 Immungene Inc MANIPULATED, MUTERATED ANTIBODY-INTERFERON FUSION MOLECULES
WO2013137869A1 (en) 2012-03-14 2013-09-19 Boehringer Ingelheim International Gmbh Combination therapy for treating hcv infection in an hcv-hiv coinfected patient population
JP2015512900A (en) 2012-03-28 2015-04-30 ベーリンガー インゲルハイム インターナショナル ゲゼルシャフト ミット ベシュレンクテル ハフツング Combination therapy to treat HCV infection in a special patient genotype subpopulation
CA2868883C (en) 2012-03-30 2022-10-04 Sorrento Therapeutics Inc. Fully human antibodies that bind to vegfr2
US9913822B2 (en) 2012-04-04 2018-03-13 Halozyme, Inc. Combination therapy with an anti-hyaluronan agent and therapeutic agent
EP3553086A1 (en) 2012-05-31 2019-10-16 Sorrento Therapeutics Inc. Antigen binding proteins that bind pd-l1
KR102172897B1 (en) 2012-06-08 2020-11-02 서트로 바이오파마, 인크. Antibodies comprising site-specific non-natural amino acid residues, methods of their preparation and methods of their use
US9315579B2 (en) 2012-06-22 2016-04-19 Sorrento Therapeutics, Inc. Antigen binding proteins that bind CCR2
WO2014004639A1 (en) 2012-06-26 2014-01-03 Sutro Biopharma, Inc. Modified fc proteins comprising site-specific non-natural amino acid residues, conjugates of the same, methods of their preparation and methods of their use
US9932565B2 (en) 2012-07-31 2018-04-03 Bioasis Technologies, Inc. Dephosphorylated lysosomal storage disease proteins and methods of use thereof
PL3584255T3 (en) 2012-08-31 2022-05-16 Sutro Biopharma, Inc. Modified amino acids comprising an azido group
US9278124B2 (en) 2012-10-16 2016-03-08 Halozyme, Inc. Hypoxia and hyaluronan and markers thereof for diagnosis and monitoring of diseases and conditions and related methods
JP2016500058A (en) 2012-11-12 2016-01-07 レッドウッド バイオサイエンス, インコーポレイテッド Methods for producing compounds and conjugates
US9310374B2 (en) 2012-11-16 2016-04-12 Redwood Bioscience, Inc. Hydrazinyl-indole compounds and methods for producing a conjugate
CN104870423A (en) 2012-11-16 2015-08-26 加利福尼亚大学董事会 Pictet-spengler ligation for protein chemical modification
US20140205566A1 (en) 2012-11-30 2014-07-24 Novartis Ag Cyclic nucleuoside derivatives and uses thereof
US9383357B2 (en) 2012-12-07 2016-07-05 Northwestern University Biomarker for replicative senescence
EP2935311B1 (en) 2012-12-20 2021-03-31 Amgen Inc. Apj receptor agonists and uses thereof
CN103908660B (en) * 2013-01-05 2015-02-04 石药集团百克(山东)生物制药有限公司 Polyethylene glycol modified rhG-CSF pharmaceutical composition and preparation method thereof
US20150361159A1 (en) 2013-02-01 2015-12-17 Bristol-Myers Squibb Company Fibronectin based scaffold proteins
CN103113466B (en) * 2013-03-01 2015-06-03 中国科学院过程工程研究所 Recombinant human interferon beta-1b modified by polyethylene glycol and preparation method of recombinant human interferon beta-1b
UY35397A (en) 2013-03-12 2014-10-31 Amgen Inc POWERFUL AND SELECTIVE INHIBITORS OF NaV1.7
BR112015022416A2 (en) 2013-03-13 2017-10-24 Bioasis Technologies Inc p97 fragments and their uses
RU2535002C2 (en) * 2013-04-04 2014-12-10 Федеральное государственное бюджетное учреждение "Научно-исследовательский институт фармакологии" Сибирского отделения Российской академии медицинских наук Method for correction of remote consequences of spermatogenesis caused by cytostatic exposure
JP2016519108A (en) 2013-04-18 2016-06-30 アルモ・バイオサイエンシーズ・インコーポレイテッド Method for using interleukin-10 for the treatment of diseases and disorders
US9717649B2 (en) 2013-04-24 2017-08-01 Corning Incorporated Delamination resistant pharmaceutical glass containers containing active pharmaceutical ingredients
US9717648B2 (en) 2013-04-24 2017-08-01 Corning Incorporated Delamination resistant pharmaceutical glass containers containing active pharmaceutical ingredients
US9707155B2 (en) 2013-04-24 2017-07-18 Corning Incorporated Delamination resistant pharmaceutical glass containers containing active pharmaceutical ingredients
US9700485B2 (en) 2013-04-24 2017-07-11 Corning Incorporated Delamination resistant pharmaceutical glass containers containing active pharmaceutical ingredients
US9839579B2 (en) 2013-04-24 2017-12-12 Corning Incorporated Delamination resistant pharmaceutical glass containers containing active pharmaceutical ingredients
US9700486B2 (en) 2013-04-24 2017-07-11 Corning Incorporated Delamination resistant pharmaceutical glass containers containing active pharmaceutical ingredients
US9603775B2 (en) 2013-04-24 2017-03-28 Corning Incorporated Delamination resistant pharmaceutical glass containers containing active pharmaceutical ingredients
US9849066B2 (en) 2013-04-24 2017-12-26 Corning Incorporated Delamination resistant pharmaceutical glass containers containing active pharmaceutical ingredients
US9707154B2 (en) 2013-04-24 2017-07-18 Corning Incorporated Delamination resistant pharmaceutical glass containers containing active pharmaceutical ingredients
US9713572B2 (en) 2013-04-24 2017-07-25 Corning Incorporated Delamination resistant pharmaceutical glass containers containing active pharmaceutical ingredients
US9707153B2 (en) 2013-04-24 2017-07-18 Corning Incorporated Delamination resistant pharmaceutical glass containers containing active pharmaceutical ingredients
JP2016528879A (en) 2013-06-17 2016-09-23 アルモ・バイオサイエンシーズ・インコーポレイテッド Methods for assessing protein identity and stability
TW201534726A (en) 2013-07-03 2015-09-16 Halozyme Inc Thermally stable PH20 hyaluronidase variants and uses thereof
WO2015006555A2 (en) 2013-07-10 2015-01-15 Sutro Biopharma, Inc. Antibodies comprising multiple site-specific non-natural amino acid residues, methods of their preparation and methods of their use
AU2014312190A1 (en) 2013-08-28 2016-02-18 Bioasis Technologies Inc. CNS-targeted conjugates of antibodies
JP6509867B2 (en) 2013-08-30 2019-05-08 アルモ・バイオサイエンシーズ・インコーポレイテッド Methods of using interleukin-10 to treat diseases and disorders
AU2014330853A1 (en) 2013-10-02 2016-02-25 National Institutes Of Health Insulin-like growth factor mimetics for use in therapy
EP3055298B1 (en) 2013-10-11 2020-04-29 Sutro Biopharma, Inc. Modified amino acids comprising tetrazine functional groups, methods of preparation, and methods of their use
WO2015057908A1 (en) 2013-10-18 2015-04-23 Novartis Ag Methods of treating diabetes and related disorders
RU2016122957A (en) 2013-11-11 2017-12-19 Армо Байосайенсиз, Инк. Methods of using interleukin-10 for the treatment of diseases and disorders
US20160296632A1 (en) 2013-11-13 2016-10-13 Aequus Biopharma, Inc. Engineered glycoproteins and uses thereof
JP6745218B2 (en) 2013-11-27 2020-08-26 レッドウッド バイオサイエンス, インコーポレイテッド Methods for producing hydrazinyl-pyrrolo compounds and conjugates
UY35874A (en) 2013-12-12 2015-07-31 Novartis Ag A PROCESS FOR THE PREPARATION OF A COMPOSITION OF PEGILATED PROTEINS
US10188739B2 (en) 2014-02-27 2019-01-29 Xenetic Biosciences, Inc. Compositions and methods for administering insulin or insulin-like protein to the brain
US10293043B2 (en) 2014-06-02 2019-05-21 Armo Biosciences, Inc. Methods of lowering serum cholesterol
CA2951391C (en) 2014-06-10 2021-11-02 Amgen Inc. Apelin polypeptides
US10106579B2 (en) 2014-06-12 2018-10-23 Ra Pharmaceuticals, Inc. Modulation of complement activity
AU2015305894A1 (en) 2014-08-22 2017-04-06 Sorrento Therapeutics, Inc. Antigen binding proteins that bind CXCR3
PL3186281T3 (en) 2014-08-28 2019-10-31 Halozyme Inc Combination therapy with a hyaluronan-degrading enzyme and an immune checkpoint inhibitor
BR112017007765B1 (en) 2014-10-14 2023-10-03 Halozyme, Inc COMPOSITIONS OF ADENOSINE DEAMINASE-2 (ADA2), VARIANTS THEREOF AND METHODS OF USING THE SAME
KR20170068553A (en) 2014-10-14 2017-06-19 아르모 바이오사이언시스 인코포레이티드 Interleukin-15 compositions and uses thereof
JP6675394B2 (en) 2014-10-22 2020-04-01 アルモ・バイオサイエンシーズ・インコーポレイテッド Use of interleukin-10 for the treatment of diseases and disorders
UY36370A (en) 2014-10-24 2016-04-29 Bristol Myers Squibb Company Una Corporación Del Estado De Delaware MODIFIED FGF-21 POLIPEPTIDES AND ITS USES
KR20230079520A (en) 2014-11-06 2023-06-07 파마에센시아 코퍼레이션 Dosage Regimen for PEGylated Interferon
US20160151511A1 (en) * 2014-12-02 2016-06-02 Antriabio, Inc. Proteins and protein conjugates with increased hydrophobicity
CN104491843B (en) * 2015-01-23 2017-09-12 石药集团百克(山东)生物制药有限公司 A kind of polyethyleneglycol modified rhG CSF active pharmaceutical compositions
US9937222B2 (en) 2015-01-28 2018-04-10 Ra Pharmaceuticals, Inc. Modulators of complement activity
WO2016126615A1 (en) 2015-02-03 2016-08-11 Armo Biosciences, Inc. Methods of using interleukin-10 for treating diseases and disorders
JP6937984B2 (en) * 2015-03-03 2021-09-22 ナンシャ・バイオロジックス・(ホンコン)・リミテッドNansha Biologics (Hong Kong) Limited Compositions and Methods of Pegated IL-11
US10711067B2 (en) 2015-03-03 2020-07-14 Xoma (Us) Llc Treatment of post-prandial hyperinsulinemia and hypoglycemia after bariatric surgery
JP6901400B2 (en) 2015-04-03 2021-07-14 ゾーマ テクノロジー リミテッド Cancer treatment using TGF-β and PD-1 inhibitors
US20180118799A1 (en) 2015-04-13 2018-05-03 National Institute Of Advanced Industrial Science And Technology Cyclized cytokine and method for producing same
CN115919996A (en) 2015-05-01 2023-04-07 雅利斯塔制药公司 Adiponectin peptidomimetics for the treatment of ophthalmic diseases
JP7121496B2 (en) 2015-05-28 2022-08-18 アルモ・バイオサイエンシーズ・インコーポレイテッド Pegylated interleukin-10 for use in cancer therapy
JP2018532693A (en) 2015-08-06 2018-11-08 ゾーマ (ユーエス) リミテッド ライアビリティ カンパニー Antibody fragments against the insulin receptor and their use to treat hypoglycemia
CN108025040A (en) 2015-08-25 2018-05-11 阿尔莫生物科技股份有限公司 The method that disease and illness are treated using interleukin-10
US10842879B2 (en) * 2015-09-18 2020-11-24 University Of Miyazaki Long-acting adrenomedullin derivative
EP3355908A1 (en) 2015-10-01 2018-08-08 Amgen Inc. Treatment of bile acid disorders
US10935276B2 (en) 2015-10-20 2021-03-02 Steven Michalski Air mixing device
WO2017100400A2 (en) 2015-12-08 2017-06-15 Biomarin Pharmaceutical Inc. Use of c-type natriuretic peptide variants to treat osteoarthritis
TWI745320B (en) 2015-12-16 2021-11-11 美商Ra製藥公司 Modulators of complement activity
CN116333124A (en) 2016-01-29 2023-06-27 索伦托药业有限公司 Antigen binding proteins that bind to PD-L1
CN109803680A (en) 2016-08-01 2019-05-24 佐马美国有限公司 (PTH1R) antibody of parathyroid hormone receptor 1 and its purposes
US10858428B2 (en) 2016-09-28 2020-12-08 Xoma (Us) Llc Antibodies that bind interleukin-2 and uses thereof
CN110087668A (en) 2016-12-07 2019-08-02 Ra制药公司 The regulator of complement activity
US11129906B1 (en) 2016-12-07 2021-09-28 David Gordon Bermudes Chimeric protein toxins for expression by therapeutic bacteria
ES2963839T3 (en) 2017-02-08 2024-04-02 Bristol Myers Squibb Co Modified relaxin polypeptides comprising a pharmacokinetic enhancer and uses thereof
CA3177086A1 (en) 2017-06-22 2018-12-27 Catalyst Biosciences, Inc. Modified membrane type serine protease 1 (mtsp-1) polypeptides and methods of use
KR102020995B1 (en) 2017-10-30 2019-09-16 한국코러스 주식회사 A method of preparing gcsf and polyol_conjugated conjugates with high yield
CN111094462A (en) 2017-12-26 2020-05-01 贝克顿·迪金森公司 Deep ultraviolet excitable water-solvated polymer dyes
US11732016B2 (en) 2017-12-27 2023-08-22 Council Of Scientific & Industrial Research Polypeptide exhibiting granulocyte-colony stimulating factor activity
CN112135837A (en) * 2018-03-14 2020-12-25 勇·巴 Pegylated antifreeze proteins and methods of making and using same
WO2019191482A1 (en) 2018-03-30 2019-10-03 Becton, Dickinson And Company Water-soluble polymeric dyes having pendant chromophores
WO2019209473A1 (en) 2018-04-24 2019-10-31 Amgen Inc. Method for making injectable pharmaceutical compositions
US20190351031A1 (en) 2018-05-16 2019-11-21 Halozyme, Inc. Methods of selecting subjects for combination cancer therapy with a polymer-conjugated soluble ph20
KR102167755B1 (en) 2018-05-23 2020-10-19 주식회사 큐어바이오 Fragmented GRS polypeptide, mutants thereof and use thereof
EP3813867A1 (en) 2018-07-22 2021-05-05 Bioasis Technologies Inc. Treatment of lymmphatic metastases
JP7441826B2 (en) 2018-09-11 2024-03-01 アンブルックス,インコーポレイテッド Interleukin-2 polypeptide conjugates and uses thereof
AU2019361206A1 (en) 2018-10-19 2021-06-03 Ambrx, Inc. Interleukin-10 polypeptide conjugates, dimers thereof, and their uses
MX2021007843A (en) 2018-12-28 2021-08-11 Vertex Pharma Modified urokinase-type plasminogen activator polypeptides and methods of use.
US11613744B2 (en) 2018-12-28 2023-03-28 Vertex Pharmaceuticals Incorporated Modified urokinase-type plasminogen activator polypeptides and methods of use
JP2022520792A (en) 2019-02-12 2022-04-01 アンブルックス,インコーポレイテッド Compositions, Methods, and Uses Containing Antibody-TLR Agonist Conjugates
US10882954B2 (en) 2019-04-11 2021-01-05 Sunbio Inc. Tertiary alkoxy polyethylene glycol and derivatives thereof
EP3955965A1 (en) * 2019-04-15 2022-02-23 NOF Corporation Conjugate of bio-related substance and block polymer, and block polymer derivative for obtaining said conjugate
EP3955957A1 (en) 2019-04-15 2022-02-23 Qwixel Therapeutics LLC Fusion protein composition(s) comprising targeted masked type i interferons (ifna and ifnb) and an antibody against tumor antigen, for use in the treatment of cancer
CA3174114A1 (en) 2020-03-11 2021-09-16 Ambrx, Inc. Interleukin-2 polypeptide conjugates and methods of use thereof
US20230204597A1 (en) 2020-03-20 2023-06-29 Amgen Inc. Determination of free n-terminus of pegfilgrastim using an acid protease
US20210355468A1 (en) 2020-05-18 2021-11-18 Bioasis Technologies, Inc. Compositions and methods for treating lewy body dementia
US20210393787A1 (en) 2020-06-17 2021-12-23 Bioasis Technologies, Inc. Compositions and methods for treating frontotemporal dementia
JP2023538071A (en) 2020-08-20 2023-09-06 アンブルックス,インコーポレイテッド Antibody-TLR agonist conjugates, methods and uses thereof
US11602598B1 (en) * 2020-08-27 2023-03-14 Fresenius Kabi Deutschland Gmbh Prefilled syringe with pegfilgrastim having optimized dose and methods related thereto
CN112710826A (en) * 2020-11-17 2021-04-27 北京九强生物技术股份有限公司 Coating and sealing method for improving stability of reagent
US11952461B2 (en) 2021-03-22 2024-04-09 Sunbio, Inc. Siloxy polyethylene glycol and derivatives thereof
CA3213805A1 (en) 2021-04-03 2022-10-06 Feng Tian Anti-her2 antibody-drug conjugates and uses thereof
CN113214328B (en) * 2021-05-08 2022-06-28 宁波经济技术开发区弘翔生化科技有限公司 Double-aqueous-phase system and monosaccharide separation method based on double-aqueous-phase system
EP4155349A1 (en) 2021-09-24 2023-03-29 Becton, Dickinson and Company Water-soluble yellow green absorbing dyes
WO2024007016A2 (en) 2022-07-01 2024-01-04 Beckman Coulter, Inc. Novel fluorescent dyes and polymers from dihydrophenanthrene derivatives
WO2024044327A1 (en) 2022-08-26 2024-02-29 Beckman Coulter, Inc. Dhnt monomers and polymer dyes with modified photophysical properties

Family Cites Families (96)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US1609546A (en) 1925-11-19 1926-12-07 Petroleum Rectifying Co Process of separating water from emulsions
DE2047413C3 (en) 1970-09-26 1980-05-29 F. Hoffmann-La Roche & Co Ag, Basel (Schweiz) Process for the production of peptides in a homogeneous phase
US4002714A (en) * 1972-08-14 1977-01-11 Fumio Usui Method for producing a tapered pipe of reinforced synthetic resin
US4179337A (en) * 1973-07-20 1979-12-18 Davis Frank F Non-immunogenic polypeptides
US4002531A (en) * 1976-01-22 1977-01-11 Pierce Chemical Company Modifying enzymes with polyethylene glycol and product produced thereby
DE2930542A1 (en) 1979-07-27 1981-02-12 Hoechst Ag NEW INSULINE DERIVATIVES AND METHOD FOR THEIR PRODUCTION
JPS57192435A (en) 1981-05-20 1982-11-26 Toyobo Co Ltd Modified polypeptide
DE3139483C2 (en) 1981-10-03 1985-06-13 Dr.-Ing. Rudolf Hell Gmbh, 2300 Kiel Method and circuit arrangement for increasing contrast
US6936694B1 (en) * 1982-05-06 2005-08-30 Intermune, Inc. Manufacture and expression of large structural genes
DE3380726D1 (en) 1982-06-24 1989-11-23 Japan Chem Res Long-acting composition
JPS58225025A (en) * 1982-06-24 1983-12-27 Nippon Chem Res Kk Long active composition
WO1985003934A1 (en) 1984-03-06 1985-09-12 Takeda Chemical Industries, Ltd. Chemically modified protein and process for its preparation
EP0154316B1 (en) * 1984-03-06 1989-09-13 Takeda Chemical Industries, Ltd. Chemically modified lymphokine and production thereof
JPS61227526A (en) 1984-07-25 1986-10-09 Chugai Pharmaceut Co Ltd Novel csf and method of collecting same
JPS6142558A (en) 1984-08-06 1986-03-01 Matsushita Electric Works Ltd Amino resin molding material
ATE65798T1 (en) 1985-02-08 1991-08-15 Chugai Pharmaceutical Co Ltd HUMAN GRANULOXCYTE COLONY STIMULATION FACTOR.
JPS62129298A (en) 1985-12-02 1987-06-11 Chugai Pharmaceut Co Ltd Novel polypeptide
US5532341A (en) 1985-03-28 1996-07-02 Sloan-Kettering Institute For Cancer Research Human pluripotent hematopoietic colony stimulating factor
DE3676670D1 (en) 1985-06-26 1991-02-07 Cetus Corp SOLUBILIZATION OF PROTEINS FOR PHARMACEUTICAL COMPOSITIONS BY POLYMER CONJUGATION.
US4766106A (en) * 1985-06-26 1988-08-23 Cetus Corporation Solubilization of proteins for pharmaceutical compositions using polymer conjugation
ES8800982A1 (en) 1985-07-05 1987-12-01 Takeda Chemical Industries Ltd Modified enzymes, production and use thereof.
JPS63500636A (en) 1985-08-23 1988-03-10 麒麟麦酒株式会社 DNA encoding multipotent granulocyte colony stimulating factor
US4810643A (en) 1985-08-23 1989-03-07 Kirin- Amgen Inc. Production of pluripotent granulocyte colony-stimulating factor
JPH0657152B2 (en) 1985-09-17 1994-08-03 中外製薬株式会社 CSF genes
JP2514950B2 (en) 1986-03-10 1996-07-10 エフ・ホフマン―ラ ロシユ アーゲー Chemically modified protein, its production method and intermediate
DK203187A (en) * 1986-04-22 1987-10-23 Immunex Corp HUMAN G-CSF PROTEIN EXPRESSION
US6673347B1 (en) 1986-04-30 2004-01-06 Gryphon Therapeutics Polypeptide and protein derivatives and process for their preparation
CA1283046C (en) 1986-05-29 1991-04-16 Nandini Katre Tumor necrosis factor formulation
US4791192A (en) 1986-06-26 1988-12-13 Takeda Chemical Industries, Ltd. Chemically modified protein with polyethyleneglycol
JPS63126900A (en) 1986-06-26 1988-05-30 Takeda Chem Ind Ltd Chemically modified protein
JPS63152393A (en) * 1986-07-03 1988-06-24 Takeda Chem Ind Ltd Glycosyl derivative
GR871067B (en) 1986-07-18 1987-11-19 Chugai Pharmaceutical Co Ltd Process for producing stable pharmaceutical preparation containing granulocyte colony stimulating factor
EP0256843A1 (en) 1986-08-11 1988-02-24 Cetus Corporation Expression of g-csf and muteins thereof and their use
JPS6360938A (en) 1986-09-02 1988-03-17 Meiji Milk Prod Co Ltd Plasminogen activator of modified tissue type and production thereof
US4894226A (en) 1986-11-14 1990-01-16 Cetus Corporation Solubilization of proteins for pharmaceutical compositions using polyproline conjugation
US5362853A (en) 1986-12-23 1994-11-08 Kyowa Hakko Kogyo Co., Ltd. Polypeptide derivatives of human granulocyte colony stimulating factor
US5214132A (en) 1986-12-23 1993-05-25 Kyowa Hakko Kogyo Co., Ltd. Polypeptide derivatives of human granulocyte colony stimulating factor
NO176799C (en) * 1986-12-23 1995-05-31 Kyowa Hakko Kogyo Kk DNA encoding a polypeptide, recombinant plasmid that encodes and can express a polypeptide and method for producing this polypeptide
US5194592A (en) 1986-12-23 1993-03-16 Kyowa Hakko Kogyo Co. Ltd. Monoclonal antibodies to novel polypeptide derivatives of human granulocyte colony stimulating factor
JPH086063B2 (en) 1987-07-22 1996-01-24 日本ペイント株式会社 Hydrophilic surface treatment agent and treatment method
US4904584A (en) * 1987-12-23 1990-02-27 Genetics Institute, Inc. Site-specific homogeneous modification of polypeptides
US4847325A (en) 1988-01-20 1989-07-11 Cetus Corporation Conjugation of polymer to colony stimulating factor-1
CA1340810C (en) * 1988-03-31 1999-11-02 Motoo Yamasaki Polypeptide derivatives of human granulocyte colony stimulating factor
JP2958019B2 (en) 1988-05-06 1999-10-06 住友製薬株式会社 Polyethylene glycol derivative, modified peptide and method for producing the same
JP2796388B2 (en) * 1988-05-13 1998-09-10 アムジエン・インコーポレーテツド Method for purifying G-CSF
US5218092A (en) 1988-09-29 1993-06-08 Kyowa Hakko Kogyo Co., Ltd. Modified granulocyte-colony stimulating factor polypeptide with added carbohydrate chains
US5349052A (en) * 1988-10-20 1994-09-20 Royal Free Hospital School Of Medicine Process for fractionating polyethylene glycol (PEG)-protein adducts and an adduct for PEG and granulocyte-macrophage colony stimulating factor
GB8824591D0 (en) * 1988-10-20 1988-11-23 Royal Free Hosp School Med Fractionation process
AU4660789A (en) 1988-11-23 1990-06-12 Genentech Inc. Polypeptide derivatives
DE68925966T2 (en) * 1988-12-22 1996-08-29 Kirin Amgen Inc CHEMICALLY MODIFIED GRANULOCYTE COLONY EXCITING FACTOR
US6166183A (en) 1992-11-30 2000-12-26 Kirin-Amgen, Inc. Chemically-modified G-CSF
EP0460101A4 (en) 1989-02-24 1992-04-15 Immunotherapeutics, Inc. Immobilized cytokines
US5324844A (en) 1989-04-19 1994-06-28 Enzon, Inc. Active carbonates of polyalkylene oxides for modification of polypeptides
US5166322A (en) 1989-04-21 1992-11-24 Genetics Institute Cysteine added variants of interleukin-3 and chemical modifications thereof
EP0648501A1 (en) * 1989-10-10 1995-04-19 Amgen Inc. Compositions comprising G-CSF for treating or preventing infections in canine and feline animals
JP2978187B2 (en) 1989-11-02 1999-11-15 日本ケミカルリサーチ株式会社 Method for producing modified superoxide dismutase
JPH04218000A (en) * 1990-02-13 1992-08-07 Kirin Amgen Inc Modified polypeptide
DE4009661C1 (en) 1990-03-26 1991-03-07 Aisa Automation Industrielle S.A., Vouvry, Ch
GB9107846D0 (en) 1990-04-30 1991-05-29 Ici Plc Polypeptides
DE4014750A1 (en) * 1990-05-08 1991-11-14 Boehringer Mannheim Gmbh MUTEINE OF THE GRANULOCYTE-STIMULATING FACTOR (G-CSF)
US5126324A (en) 1990-06-07 1992-06-30 Genentech, Inc. Method of enhancing growth in patients using combination therapy
IE912365A1 (en) * 1990-07-23 1992-01-29 Zeneca Ltd Continuous release pharmaceutical compositions
AU660633B2 (en) * 1990-10-17 1995-07-06 Amgen, Inc. Methods and compositions for the treatment of cell proliferation disorders
US5372808A (en) * 1990-10-17 1994-12-13 Amgen Inc. Methods and compositions for the treatment of diseases with consensus interferon while reducing side effect
US5252714A (en) * 1990-11-28 1993-10-12 The University Of Alabama In Huntsville Preparation and use of polyethylene glycol propionaldehyde
US5124297A (en) 1990-12-07 1992-06-23 Amoco Corporation Olefin polymerization and copolymerization catalyst
CH682636A5 (en) * 1990-12-21 1993-10-29 Bucher Guyer Ag Masch A method for the selective removal of sugar from beverages.
DE69230789T3 (en) 1991-01-18 2007-10-31 Amgen Inc., Thousand Oaks METHODS OF TREATING ILLNESSES RELEASED BY THE TUMOR NEKROSE FACTOR
DE4105480A1 (en) 1991-02-21 1992-08-27 Boehringer Mannheim Gmbh IMPROVED ACTIVATION OF RECOMBINANT PROTEINS
ATE240740T1 (en) 1991-03-15 2003-06-15 Amgen Inc PEGYLATION OF POLYPEPTIDES
US5595732A (en) 1991-03-25 1997-01-21 Hoffmann-La Roche Inc. Polyethylene-protein conjugates
WO1993000109A1 (en) 1991-06-28 1993-01-07 Genentech, Inc. Method of stimulating immune response using growth hormone
US5281698A (en) 1991-07-23 1994-01-25 Cetus Oncology Corporation Preparation of an activated polymer ester for protein conjugation
US5197592A (en) * 1991-08-05 1993-03-30 Fmc Corporation Wire frame idler roll support
NZ244778A (en) * 1991-10-21 1994-03-25 Ortho Pharma Corp Peg imidates and protein derivatives thereof
US5589365A (en) * 1991-11-29 1996-12-31 Banyu Pharmaceutical Co., Ltd. Process for producing glycosylated indolopyrrolocarbazole derivatives by culturing certain microorganisms
JP3235855B2 (en) 1991-12-19 2001-12-04 住友製薬株式会社 Cell adhesion active peptide and its modified polymer
JP3387519B2 (en) * 1992-03-04 2003-03-17 株式会社日立製作所 Information recording / reproducing method and disk-shaped recording medium
FR2692736B1 (en) 1992-06-23 1996-12-20 Thomson Csf HIGH AND MEDIUM POWER RADIOELECTRIC FILTER.
US5382657A (en) * 1992-08-26 1995-01-17 Hoffmann-La Roche Inc. Peg-interferon conjugates
WO1994012219A2 (en) 1992-11-25 1994-06-09 Amgen Boulder Inc. Modified insulin-like growth factors
US5581476A (en) 1993-01-28 1996-12-03 Amgen Inc. Computer-based methods and articles of manufacture for preparing G-CSF analogs
AU7113594A (en) * 1993-06-21 1995-01-17 Enzon, Inc. Site specific synthesis of conjugated peptides
US5589356A (en) 1993-06-21 1996-12-31 Vanderbilt University Litigation of sidechain unprotected peptides via a masked glycoaldehyde ester and O,N-acyl rearrangement
US5481571A (en) 1993-11-12 1996-01-02 Pacific Communication Sciences, Inc. Method and apparatus for switching between radio frequency circuits
HU218893B (en) * 1994-03-31 2000-12-28 Amgen Inc. Water soluble compositions and methods for stimulating megakaryocyte growth and differentiation
US5795569A (en) * 1994-03-31 1998-08-18 Amgen Inc. Mono-pegylated proteins that stimulate megakaryocyte growth and differentiation
US5646113A (en) 1994-04-07 1997-07-08 Genentech, Inc. Treatment of partial growth hormone insensitivity syndrome
US5935924A (en) 1994-04-15 1999-08-10 Genentech, Inc. Treatment of congestive heart failure
US5661122A (en) 1994-04-15 1997-08-26 Genentech, Inc. Treatment of congestive heart failure
JPH10500693A (en) 1994-05-24 1998-01-20 アムジエン・ブルダー・インコーポレーテツド Modified insulin-like growth factor
US5824784A (en) * 1994-10-12 1998-10-20 Amgen Inc. N-terminally chemically modified protein compositions and methods
US5770577A (en) * 1994-11-14 1998-06-23 Amgen Inc. BDNF and NT-3 polypeptides selectively linked to polyethylene glycol
JP3708151B2 (en) 1994-12-15 2005-10-19 協和醗酵工業株式会社 Quantification of PEGylated human granulocyte colony-stimulating factor
US5672662A (en) 1995-07-07 1997-09-30 Shearwater Polymers, Inc. Poly(ethylene glycol) and related polymers monosubstituted with propionic or butanoic acids and functional derivatives thereof for biotechnical applications
KR101441331B1 (en) 2013-07-19 2014-09-17 주식회사 슈프리마 Optical type fingerprint recognition apparatus

Also Published As

Publication number Publication date
JPH0925298A (en) 1997-01-28
JP3177251B2 (en) 2001-06-18
US8258262B2 (en) 2012-09-04
IL134754A (en) 2009-11-18
DE69509628D1 (en) 1999-06-17
US20130189219A1 (en) 2013-07-25
CA2307142C (en) 2010-09-21
ATE277078T1 (en) 2004-10-15
ES2131811T3 (en) 1999-08-01
NL300106I2 (en) 2003-02-03
NL300106I1 (en) 2003-01-06
LU91006I2 (en) 2003-03-31
ZA951008B (en) 1996-10-18
CA2178752A1 (en) 1996-04-25
MX9602259A (en) 1997-02-28
ES2224197T3 (en) 2005-03-01
EP0822199A2 (en) 1998-02-04
CA2178752C (en) 2000-10-17
IL134754A0 (en) 2001-04-30
DE10299044I1 (en) 2003-03-27
CN101381409A (en) 2009-03-11
JP3177449B2 (en) 2001-06-18
US7090835B2 (en) 2006-08-15
EP0733067A1 (en) 1996-09-25
JP2006045243A (en) 2006-02-16
CN1071760C (en) 2001-09-26
DE69533556D1 (en) 2004-10-28
EP2392594A1 (en) 2011-12-07
CN1313343A (en) 2001-09-19
CN1229388C (en) 2005-11-30
US7662933B2 (en) 2010-02-16
AU1841995A (en) 1996-05-06
US20100310510A1 (en) 2010-12-09
CN101381409B (en) 2013-03-27
KR100248111B1 (en) 2000-03-15
DK0733067T3 (en) 1999-11-01
JP5350330B2 (en) 2013-11-27
EP0733067B1 (en) 1999-05-12
EP1564219A1 (en) 2005-08-17
CN1896103A (en) 2007-01-17
KR100261030B1 (en) 2000-06-15
JPH09506116A (en) 1997-06-17
CN1139932A (en) 1997-01-08
PT822199E (en) 2005-01-31
DE69509628T2 (en) 1999-09-16
US20060233746A1 (en) 2006-10-19
US20040181035A1 (en) 2004-09-16
JP2003327600A (en) 2003-11-19
IL112585A0 (en) 1995-05-26
NZ281469A (en) 1997-04-24
HK1008787A1 (en) 1999-05-21
WO1996011953A1 (en) 1996-04-25
EP0822199B1 (en) 2004-09-22
EP0822199A3 (en) 2001-10-24
ATE179991T1 (en) 1999-05-15
US5985265A (en) 1999-11-16
GR3030526T3 (en) 1999-10-29
US20120296072A1 (en) 2012-11-22
IL112585A (en) 2000-08-31
DE69533556T2 (en) 2005-10-20
EP2399930A1 (en) 2011-12-28
JP2006077021A (en) 2006-03-23
CA2307142A1 (en) 1996-04-25
JPH11310600A (en) 1999-11-09
AU706700B2 (en) 1999-06-24
HK1008826A1 (en) 1999-05-21
JP2010215657A (en) 2010-09-30
US5824784A (en) 1998-10-20
JP2003155299A (en) 2003-05-27

Similar Documents

Publication Publication Date Title
CA2307142C (en) N-terminally chemically modified protein compositions and methods
US6956027B2 (en) N-terminally chemically modified protein compositions and methods
AU741659B2 (en) N-terminally chemically modified protein compositions and methods
Gabriel et al. United States Patent (19)
AU2004235682B2 (en) N-Terminally chemically modified protein compositions and methods
MXPA96000206A (en) Compositions and methods to stimulate the growth and differentiation of the megacarioci

Legal Events

Date Code Title Description
EEER Examination request
FZDE Discontinued