WO1987000056A1 - Solubilization of proteins for pharmaceutical compositions using polymer conjugation - Google Patents

Solubilization of proteins for pharmaceutical compositions using polymer conjugation Download PDF

Info

Publication number
WO1987000056A1
WO1987000056A1 PCT/US1986/001252 US8601252W WO8700056A1 WO 1987000056 A1 WO1987000056 A1 WO 1987000056A1 US 8601252 W US8601252 W US 8601252W WO 8700056 A1 WO8700056 A1 WO 8700056A1
Authority
WO
WIPO (PCT)
Prior art keywords
protein
peg
pegylated
polymer
group
Prior art date
Application number
PCT/US1986/001252
Other languages
English (en)
French (fr)
Inventor
Nandini Katre
Michael J. Knauf
Original Assignee
Cetus Corporation
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Cetus Corporation filed Critical Cetus Corporation
Priority to AT86903990T priority Critical patent/ATE59301T1/de
Priority to AU59700/86A priority patent/AU580431B2/en
Priority to DE8686903990T priority patent/DE3676670D1/de
Priority to IN464/CAL/86A priority patent/IN163200B/en
Publication of WO1987000056A1 publication Critical patent/WO1987000056A1/en
Priority to DK097987A priority patent/DK169874B1/da
Priority to NO870779A priority patent/NO171295C/no
Priority to FI870809A priority patent/FI93424C/fi

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K1/00General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length
    • C07K1/107General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length by chemical modification of precursor peptides
    • C07K1/1072General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length by chemical modification of precursor peptides by covalent attachment of residues or functional groups
    • C07K1/1077General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length by chemical modification of precursor peptides by covalent attachment of residues or functional groups by covalent attachment of residues other than amino acids or peptide residues, e.g. sugars, polyols, fatty acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/19Cytokines; Lymphokines; Interferons
    • A61K38/20Interleukins [IL]
    • A61K38/2013IL-2
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/19Cytokines; Lymphokines; Interferons
    • A61K38/21Interferons [IFN]
    • A61K38/215IFN-beta
    • 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
    • A61K47/30Macromolecular organic or inorganic compounds, e.g. inorganic polyphosphates
    • A61K47/42Proteins; Polypeptides; Degradation products thereof; Derivatives thereof, e.g. albumin, gelatin or zein
    • 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
    • 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
    • A61K47/68Medicinal 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 antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6801Drug-antibody or immunoglobulin conjugates defined by the pharmacologically or therapeutically active agent
    • A61K47/6803Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates
    • A61K47/6811Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates the drug being a protein or peptide, e.g. transferrin or bleomycin
    • A61K47/6817Toxins
    • A61K47/6819Plant toxins
    • A61K47/6825Ribosomal inhibitory proteins, i.e. RIP-I or RIP-II, e.g. Pap, gelonin or dianthin
    • A61K47/6827Ricin A

Definitions

  • This invention relates to a chemical modification of biologically active proteins which alters the chemical and/or physiological properties of these proteins. More specifically, this invention relates to selective conjugation of lipophilic water- insoluble proteins to polymers to render the proteins soluble at physiological pH.
  • heterologous proteins produced in icrobial host cells are found as insoluble material in refractile bodies.
  • heterologous proteins which form refractile bodies in commonly found culture conditions include interleu in-2 (IL-2), interferon- ⁇ (IFN- ⁇ ), feline leukemia virus (FeLV) envelope protein, human growth hormone (hGH), bovine growth hormone (bGH), porcine growth hormone (pGH), and certain proteins coated or fused with a virus such as FMD virus.
  • IL-2 interleu in-2
  • IFN- ⁇ interferon- ⁇
  • FeLV feline leukemia virus
  • hGH human growth hormone
  • bGH bovine growth hormone
  • porcine growth hormone pGH
  • certain proteins coated or fused with a virus such as FMD virus such as FMD virus.
  • many of these proteins are hydrophobic in nature and tend to stick to materials and to themselves (i.e., aggregate) rather than remain in solution.
  • many of these recombinant proteins are unglycosylated, whereas their native
  • Modifications of these proteins which might alter their solubility properties would be desirable to increase production yields of these proteins as well as to facilitate their formulation for therapeutic use.
  • modifications may reduce or eliminate aggregation of the protein when it is introduced in vivo, thereby reducing its immunogenicity.
  • polypeptides in circulatory systems for the purpose of producing a particular physiological response is well known in the medicinal arts.
  • a limitation to the potential therapeutic benefit derived from the clinical use of polypeptides is their ability to elicit an immune response in the circulatory system. This immune response may be caused by aggregates in the material prior to injection as described by R. Illig (1970), J. Cl in. Endrocr., 31, 679- 688, W. Moore (1978), J. Clin. Endrocr i no! . Metab., 46, 20-27 and W. Moore and P. Leppert (1980), J. Clin. Endrocrinol. Metab., 51, 691- 697.
  • This response involves the production of antibodies to the polypeptides by the circulatory system into which they are injected.
  • This antibody production may decrease or eliminate the desired biological function of the polypeptide, sometimes by causing reduced residence time in the circulatory system (reduced half-life) or by modifying the molecule by virtue of the antibody-polypeptide interaction.
  • 4,301,144 describes conjugation of hemoglobin to PEG, PPG, a copoly er of ethylene glycol with propylene glycol, or ethers, esters or dehydrated products of such polymers to increase the oxygen- carrying ability of the hemoglobin molecule.
  • European Patent Publication 98,110 published January 11, 1984, discloses that conjugating of a polypeptide or glycoprotein to a polyoxyethylene- polyoxypropylene copolymer increases the length of its physiological activity.
  • the polypeptide or glycoprotein is an enzyme or native interferon, which are water soluble.
  • 4,179,337 discloses conjugating of water-soluble polypeptides such as enzymes and insulin to PEG or PPG to reduce the immunogenicity of the polypeptide while retaining a substantial proportion of its desired physiological activity.
  • U.S. Patent No. 4,002,531 discloses a different method of conjugating enzymes to PEG through an aldehyde der vative.
  • U.S. Patent 4,055,635 discloses pharmaceutical compositions comprising a water-soluble complex of a proteolytic enzyme linked covalently to a polymeric substance such as polysaccharides.
  • U.S. Patent 3,960,830 discloses peptides bound to a polyalkylene glycol polymer such as polyethylene glycol.
  • U.S. Patent 4,088,538 discloses a reversibly soluble enzymatically active polymer enzyme product comprising an enzyme covalently bonded to an organic polymer such as polyethylene glycol.
  • U.S. Patent 4,415,665 discloses a method of conjugating an organic ligand containing at least one primary or secondary amino group, at least one thiol group and/or at least one aromatic hydroxy group (described in col. 3, lines 19-36) to a polymeric carrier with at least one hydroxyl group (described in col. 2, lines 42-66).
  • U.S. Patent 4,495,285 discloses a non-immunogen c plas inogen activator, the amino acid side chains of which are coupled to a polyalkylene glycol through a coupling agent.
  • U.S. Patent 4,412,989 discloses an oxygen-carrying material containing hemoglobin or a derivative thereof covalently coupled through an amide bond to polyethylene or polypropylene glycol.
  • U.S. Patent 4,496,689 discloses a covalently attached complex of alpha-1-proteinase inhibitor with a polymer such as PEG or met ho x polyethylene glycol s.
  • U.S. Patent 3,619,371 discloses a polymeric matrix having a biologically active substance chemically bound thereto.
  • U.S. Patent 3,788,948 discloses use of organic cyanate compounds to bind proteins to polymers.
  • U.S. Patent 3,876,501 discloses activation of water-soluble carbohydrates with cyanogen bromide to improve their binding to enzymes and other proteins.
  • U.S. Patent 4,055,635 discloses pharmaceutical compositions of a proteolytic enzyme linked covalently to a polymeric substance.
  • EP 152,847 discloses an enzyme conjugate composition comprising an enzyme conjugate, a calcium salt, and a polyethylene glycol.
  • JP 5792435 published November 26, 1982 discloses modified polypeptides where all or part of the amino groups are substituted with a polyethoxyl moiety.
  • DE 2312615 published September 27, 1973 discloses conjugating of polymers to compounds containing hydroxy or amino groups.
  • EP 147,761 discloses a covalent conjugate of alpha-1- proteinase inhibitor and water-soluble polymer, where the polymer may be polyethylene glycol.
  • U.S. Patent No. 4,414,147 describes rendering interferon less hydrophobic by conjugating it to an anhydride of a dicarboxylic acid such as poly(ethylene succinic anhydride).
  • EP 154,316 published September 11, 1985 to Takeda Chemical Industries, Ltd., discloses and claims chemically modified lymphokines such as IL-2 containing PEG bonded directly to at least one primary amino group of a lymphokine.
  • the present invention provides for modifying those proteins selected from ⁇ -interferon, interleukin-2, and im unotoxins which are not ordinarily soluble in water under ambient conditions at pharmaceutically acceptable pH ranges to render them soluble in aqueous buffer under such conditions.
  • This modification may be mimicking glycosylation of the protein, thereby surprisingly rendering the protein soluble as the native glycosylated protein is soluble.
  • This modification also avoids addition of extraneous solubilizing additives such as detergents or denaturants to keep the protein in solution.
  • the modified protein retains the biological activity of the unmodified protein, both initially and over time.
  • the modification under some conditions increases the physiological half-life of the protein and may decrease its immunogenicity by reducing or eliminating aggregation of the protein or by masking antigenic determinants. It has also been found that this prolonged half-life is related to the efficacy of the protein.
  • the _in_ vivo half-life can be modulated by selecting appropriate conditions and polymers.
  • the present invention is directed to a pharmaceutical composition
  • a pharmaceutical composition comprising a non-toxic, inert, pharmaceutically acceptable aqueous carrier medium in which is dissolved a biologically active selectively conjugated protein selected from the group consisting of ⁇ -interferon, interleukin-2, and an immunotoxin, wherein the protein is covalently conjugated to a water-soluble polymer selected from the group consisting of polyethylene glycol homopolymers and polyoxyethylated polyols, wherein said homopolymer is unsubstituted or substituted at one end with an alkyl group, and said polyol is unsubstituted, and wherein said protein in its unconjuga ed form is normally hydrophobic and not soluble in said aqueous carrier medium at pH 6-8 in the absence of a solubilizing agent.
  • the polymer is unsubstituted polyethylene glycol
  • PEG polymethyl PEG
  • PEG polyoxyethylated glycerol
  • Another aspect of this invention resides in a process for preparing a pharmaceutical composition comprising:
  • FIG. 1 shows densito etry scans of 14% SDS-polyacrylamide gels for molecular weight analysis of PEG-modified (PEGylated) IL-2 obtained from reactions at 0, 10, 20, 50 and 100 moles activated PEG (PEG*) per mole of IL-2.
  • Figure 2 illustrates the solubility of PEGylated IL-2 compared to unmodified IL-2 at two different pHs by absorbance scan from 200 to 650 nm.
  • Figure 3 depicts the pharmacokinetics of PEGylated and unmodified IL-2 after intravenous injection into mice.
  • Figure 4 shows the pharmacokinetics of PEGylated and unmodified IL-2 after subcutaneous injection into mice.
  • Figure 5 shows densitometry scans of 14% nonreducing SDS- polyacrylamide gels for molecular weight analysis of PEGylated IFN- ⁇ obtained from reactions at 0, 10, 20 and 50 moles PEG per mole of IFN- ⁇ .
  • Figure 6 illustrates the solubility of PEGylated IFN- ⁇ compared to unmodified IFN- ⁇ at two different pHs by absorbance scan from 200 to 650 nm.
  • the term "normally hydrophobic, water insoluble” as describing the proteins refers to those proteins which are insoluble or not readily soluble in water or an aqueous medium under ambient conditions of room temperature and atmospheric pressure at a pH of between about 6 and 8, i.e, at about neutral or physiological pH.
  • the modification herein acts to increase the solubility of such proteins when they are subjected to such physiological conditions.
  • solubility may be tested by (1) turbidity, as measured by spectrophotometric means, (2) S va.lue, as measured by ultracentrifugation, wherein the monomeric protein sedimentation rate rather than the much greater aggregate sedimentation rate signals solubility, and (3) apparent native molecular weight, as measured by size exclusion chromatography, wherein the soluble protein is closer to this value than the insoluble protein.
  • turbidity as measured by spectrophotometric means
  • S va.lue as measured by ultracentrifugation
  • apparent native molecular weight as measured by size exclusion chromatography
  • interferon- ⁇ and interleukin-2 herein may be obtained from tissue cultures or by recombinant techniques, and from any mammalian source such as, e.g., mouse, rat, rabbit, primate, pig, and human.
  • mammalian source such as, e.g., mouse, rat, rabbit, primate, pig, and human.
  • proteins are derived from a human source, and more preferably are recombinant, human proteins.
  • IFN- ⁇ recombinant ⁇ -interferon
  • fibroblast interferon having comparable biological activity to native IFN- ⁇ prepared by recombinant
  • the gene coding for interferon is excised from its native plasmid and inserted into a cloning vector to be cloned and then into an expression vector, which is used to transform a host organism, preferably a microorganism, and most preferably _E ⁇ coli.
  • the host organism expresses the foreign interferon gene under certain conditions to produce IFN- ⁇ . More preferably, the IFN- ⁇ is a utein as described in U.S. Patent No.
  • the IFN- ⁇ mutein is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-a neutral amino acid such as serine or alanine.
  • IL-2 recombinant interleukin-2
  • IL-2 preferably human IL-2
  • IL-2 refers to interleukin-2 having comparable biological activity to native IL-2 prepared by recombinant DNA techniques as described, e.g., by Taniguchi et al., Nature, 302:305- 310.(1983) and Devos, Nucleic Acids Research, 11:4307-4323 (1983).
  • the gene coding for IL-2 is excised from its native plas id and inserted into a cloning vector to be cloned and then into an expression vector, which is used to transform a host organism, preferably a microorganism, and most preferably E ⁇ col .
  • the host organism expresses the foreign gene to produce IL-2 under expression conditions.
  • the IL-2 is a mutein as described in U.S. Patent No. 4,518,584, in which the cysteine normally occurring at position 125 of the wild-type or native molecule has been replaced by a neutral amino acid such as serine or alanine.
  • the IL-2 mutein may be one in which the methionine normally occurring at position 104 of the wild-type or native molecule has been replaced by a neutral amino acid such as alanine.
  • the IL-2 is a protein produced by a microorganism or by yeast which has been transformed with the human cDNA sequence of IL-2 which encodes a protein with an amino acid sequence at least substantially identical to the amino acid sequence of native human IL-2, including the disulfide bond of the cysteines at positions 58 and 105, and has biological activity which is common to native human IL-2.
  • Substantial identity of amino acid sequences means the sequences are identical or differ by one or more amino acid alterations (deletions, additions, substitutions) which do not cause an adverse functional dissimilarity between the synthetic protein and native human IL-2.
  • IL-2 proteins with such properties include those described by Taniguchi et al., supra; Devos, supra; European Patent Publication Nos. 91,539 and 88,195; U.S. Patent 4,518,584, supra.
  • the IL-2 is ser ⁇ 1 ---.?, des- ala ⁇ seri25l-L-2, des-ala j IL-2, des-ala ⁇ ala ⁇ o4lL-2, or des- ala j alai Q 4 ser l25.lL--.2 » where "des-ala ⁇ " indicates that the N-terminal alanyl residue of the IL-2 has been deleted.
  • the precise chemical structure of the proteins herein will depend on a number of factors.
  • a particular protein may be obtained as an acidic or basic salt, or in neutral form. All such preparations which retain their bioactivity when placed in suitable environmental conditions are included in the definition of proteins herein.
  • the primary amino acid sequence of the protein may be augmented by derivatization using sugar moieties (glycosylation) or by other supplementary molecules such as lipids, phosphate, acetyl groups and the like, more commonly by conjugation with saccharides. Certain aspects of such augmentation are accomplished through post- translational processing systems of the producing host; other such modifications may be introduced _in_ vitro. In any event, such modifications are included in the definition of protein herein so long as the bioactivity of the protein is not destroyed. It is expected, of course, that such modifications may quantitatively or qualitatively affect the bioactivity by either enhancing or diminishing the activity of the protein in the various assays.
  • hydrophobic recombinant proteins such as IL-2 and IFN- ⁇ produced from transformed host cells containing recombinant DNA precipitate inside the cell as opposed to being soluble in the cell culture medium.
  • the intracellularly produced protein must be separated from the cellular debris and recovered from the cell before it can be formulated into a purified biologically active material.
  • the cell membrane of the transformed host microorganism is disrupted, greater than 99% by weight of the salts is removed from the disruptate, the desalted disruptate is redisrupted, a material, preferably a sugar such as sucrose, is added to the disruptate to create a density or viscosity gradient in the liquid within the disruptate, and the refractile material is separated from the cellular debris by high-speed centrifugation, i.e., at about 10,000 to 40,000 x g.
  • the salts are removed from the disruptate by diafiltration or centrifugation and sucrose is added to increase the density of the liquid to about 1.1 to 1.3 g/ml.
  • the pellet containing the refractile bodies is solubilized with a denaturant such as sodium dodecyl sulfate, the resulting suspension is centrifuged, and the supernate containing the protein is treated to isolate the protein.
  • the protein is separated from the supernate by appropriate means such as reverse-phase high pressure liquid chromatography (RP-HPLC) and/or gel filtration chromatography. After such separation, the protein is preferably oxidized to ensure the production of high yields of recombinant protein in a configuration most like its native counterpart.
  • RP-HPLC reverse-phase high pressure liquid chromatography
  • gel filtration chromatography gel filtration chromatography
  • the oxidation may also be carried out by reacting an aqueous solution containing a solubilized form of the protein at a pH between about 5.5 and 9 in the presence of air with at least an effective amount of an oxidation promoter containing a Cu + cation, as described in U.S. Patent No. 4,572,798 to . Koths et al.
  • the preferred oxidation promoter or oxidant is CuCl 2 or (o- phenanthroline)2 Cu .
  • the protein may optionally be desalted and purified further by RP-HPLC, dilution/diafiltration, S- 200 gel filtration chromatography, and ultrafiltration techniques before modification with activated polymer as described further hereinbelow.
  • the polymer modification may, however, be carried out at any step after the heterologous protein has been isolated in sufficiently pure form to be biologically active for therapeutic purposes. The point at which the modification will occur will depend on the ultimate purity of the protein required for the final pharmaceutical formulation and use.
  • immunotoxin refers to a conjugate of an antibody and a cytotoxic moiety.
  • the cytotoxic moiety of the immunotoxin includes a cytotoxic drug or an enzymatically active toxin of bacterial or plant origin or an enzymatically active fragment ("A chain") of such a toxin.
  • Examples of enzymatically active toxins and fragments thereof include diphtheria A chain, nonbinding active fragments of diphtheria toxin, exotoxin A chain (from Pseudomonas aeruginosa), ricin A chain, abrin A chain, odeccin A chain, alpha-sarcin, Aleurites fordii proteins, dianthin proteins, Ph tolacca ameri cana proteins (PAPI , PAPII , and PAP-S ), momordica charantia inhibitor, curcin, crotin, saponaria officinalis inhibitor, gelonin, itogell in, restrictocin, phenomycin, and eno ycin.
  • Ricin A chain , nonbinding active fragments of diphtheria toxin, abrin A chain, and PAPII are preferred. Most preferred is the ricin A chain, which is modified by reaction with the polymer.
  • the antibodies employed in the immunotoxin are preferably monoclonal antibodies directed against a speci fi c pathologi cal condition such as, e.g. , cancers such as breast, prostate, colon or ovarian cancer , melanoma , myeloma , etc.
  • Conjugates of the antibody and cytotoxic moiety may be made using a variety of bifunctional protein modifying reagents .
  • reagents include N-succinimidyl -3-(2-pyridyldi thio) propi onate (SPDP), iminothiolate (IT) , bi functional derivatives of i idoesters such as dimethyl adipimidate * HC1 , active esters such as di succinim dyl suberate, aldehydes such as gl utaraldehyde, bis-azido compounds such as bis (p-azidobenzoyl ) hexanediamine, bis-diazonium derivatives such as bis-(p-dia zoni um-benzoyl )-ethyl enediamine , di isocyanates such as tolylene-2,6-diisocyanate, and bis-active fluorine compounds such as l ,5
  • residues may be any reactive amino acids on the protein, such as one or two cysteines or the N-terminal amino acid group, preferably the reactive amino acid is lysine, which is l inked to the reactive group of the activated polymer through its free ⁇ -amino group, or glutamic or asparti c aci d, which is l inked to the polymer through an amide bond.
  • the protein is covalently bonded via one or two of the amino acid residues of the protein , preferably lysines , for maximum biological activity.
  • the protein is covalently bonded via up to ten of the amino acid residues of the protein, preferably lysines, with higher substitutions generally increasing the circulatory life of the protein.
  • the three types of proteins described above which are normally hydrophobic and water insoluble, are rendered soluble in an aqueous carrier medium, preferably at a pH of about 5 to 8, more preferably about 6-8 and most preferably, 6.5-7.8, without use of solubilizing agents, by modifying the proteins through conjugation to a specified polymer.
  • the pH of the reaction is preferably about 7 to 9, more preferably 8-9.
  • the success of such a modification of these proteins cannot be predicted from earlier use of polymer modification of water-soluble enzymes and hormones.
  • the polymer to which the protein is attached is a homopolymer of polyethylene glycol (PEG) or is a polyoxyethylated polyol, provided in all cases that the polymer is soluble in water at room temperature.
  • PEG polyethylene glycol
  • polyoxyethylated polyol include, for example, polyoxyethylated glycerol , polyoxyethylated sorbitol, polyoxyethylated glucose, or the like.
  • the glycerol backbone of polyoxyethylated glycerol is the same backbone occurring naturally in, for example, animals and humans in mono-, di-, and triglycerides. Therefore, this branching would not necessarily be seen as a foreign agent in the body.
  • the polymer need not have any particular molecular weight, but it is preferred that the molecular weight be between about 300 and 100,000, more preferably between 350 and 40,000, depending, for example, on the particular protein employed.
  • the PEG homopolymer is unsubstituted, but it may also be substituted at one end with an alkyl group.
  • the alkyl group is a C1-C4 alkyl group, and most preferably a methyl group.
  • the polymer is an unsubstituted homopolymer of PEG, a monomethyl -substituted homopolymer of PEG or polyoxyethylated glycerol, and has a molecular weight of about 350 to 40,000.
  • the protein is conjugated via a terminal reacti ve group on the polymer.
  • the polymer with the reactive group(s) is designated herein as activated polymer.
  • the reactive group selectively reacts with free amino or other reactive groups on the protei n. It wi l l be understood, however, that the type and amount of the reactive group chosen, as well as the type of polymer employed, to obtain optimum results, wil l depend on the protein employed to avoid having the reactive group react with too many particularly active groups on the protein. As this may not be possible to avoid completely, it is recommended that general ly from about 0.1 to 1000 moles , preferably 2- 200 moles, of acti vated polymer per mole of protein, dependi ng on protein concentration, is employed.
  • the amount of acti vated polymer employed is preferably no more than 50 moles per mole of IL-2, and most preferably i s about 2 to 20 mol es per mol e of IL-2, depending on the speci fi c properties ul timately desired, i .e., the final amount is a balance to maintain optimum activity , while at the same time optimizing, if possibl e, the half-l ife of the protein.
  • at l east about 50% of the biological activity of the protein is retained, and most preferably 100% is retained.
  • the covalent modi fication reaction may take place by any suitable method generally used for reactive biological ly acti ve material s with inert polymers , preferably at about pH 5-9, i f the reacti ve groups on the protein are lysine groups .
  • general ly the process invol ves preparing an activated polymer (with at least one terminal hydroxyl group) and thereafter reacting the protein with the activated polymer to produce the sol ubil i zed protein suitabl e for formulation.
  • the above modi fication reaction can be performed by several methods, which may involve one or more steps.
  • suitabl e modi fying agents which can be used to produce the acti vated polymer in a one-step reaction incl ude cyanuri c aci d chloride (2,4,6-t ⁇ ' chloro-S- triazine) and cyanuric acid fluoride.
  • the modification reaction takes place in two steps wherein the polymer is reacted first with an acid anhydride such as succinic or glutaric anhydride to form a carboxylic acid, and the carboxylic acid is then reacted with a compound capable of reacting with the carboxylic acid to form an activated polymer with a reactive ester group which is capable of reacting with the protein.
  • an acid anhydride such as succinic or glutaric anhydride
  • a compound capable of reacting with the carboxylic acid to form an activated polymer with a reactive ester group which is capable of reacting with the protein.
  • examples of such compounds include N-hydroxysuccinimide, 4- hydroxy-3-nitrobenzene sulfonic acid, and the like, and preferably N- hydroxysuccinimide or 4-hydroxy-3-nitrobenzene sulfonic acid is used.
  • monomethyl substituted PEG may be reacted at elevated temperatures, preferably about 100-110°C for four hours, with glutaric anhydride.
  • the monomethyl PEG-glutaric acid thus produced is then reacted with N-hydroxysuccinimide in the presence of a carbodiimide reagent such as dicyclohexyl or isopropyl carbodii ide to produce the activated polymer, methoxypolyethylene glycolyl-N- succinimidyl glutarate, which can then be reacted with the protein.
  • a carbodiimide reagent such as dicyclohexyl or isopropyl carbodii ide
  • the monomethyl substituted PEG may be reacted with glutaric anhydride followed by reaction with 4-hydroxy-3-nitrobenzene sulfonic acid (HNSA) in the presence of dicyclohexyl carbodiimide to produce the activated polymer.
  • HNSA 4-hydroxy-3-nitrobenzene sulfonic acid
  • ester bonds are chemically and physiologically less stable than amide bonds, it . may be preferable to use chemical transformations in the conjugating reaction which would produce carboxylic acids or amides without concurrent production of esters.
  • the protein thus modified is then formulated in a non-toxic, inert, pharmaceutically acceptable aqueous carrier medium, preferably at a pH of about 3 to 8, more preferably 6-8.
  • aqueous formulations compatible with the culture or perfusion medium will generally be used.
  • the sterile product When used in vivo for therapy, the sterile product will- consist of a mixture of protein dissolved in an aqueous buffer in an amount which will provide a pharmaceutically acceptable pH when the mixture is reconstituted.
  • a water-soluble carrier such as mannitol may optionally be added to the medium.
  • the currently formulated unmodified IL-2 is stable for at least six months at 4°C.
  • the dosage level of protein in the formulation will depend on the in vivo efficacy data obtained after preclinical testing and will depend mainly on the protein employed and ultimate use.
  • the lyophilized mixture may be reconstituted by injecting into the vial a conventional parenteral aqueous injection such as, e.g., distilled water.
  • the reconstituted formulation prepared as described above is suitable for parenteral administration to humans or other mammals in therapeutically effective amounts (i.e., amounts which eliminate or reduce the patient's pathological condition) to provide therapy thereto, the type of therapy being dependent on the type of protein.
  • IL-2 therapy is appropriate for a variety of immunomodulatory indications such as T cell mutagenesis, induction of cytotoxic T cells, augmentation of natural killer cell activity, induction of IFN-gamma, restoration or enhancement of cellular immunity (e.g., treatment of immune deficient conditions), and augmentation of cell mediated anti-tumor activity.
  • the IL-2 may be administered in an adoptive immunotherapy method, together with isolated, lymphokine-activated lymphocytes, in a pharmaceutically acceptable carrier, where the lymphocytes are reactive to tumor when administered with the IL-2 to humans suffering from the tumor.
  • IFN- ⁇ therapy is appropriate for anti-cancer, anti-viral and anti-psoriasis treatment.
  • Specific cancers against which IFN- ⁇ has shown some efficacy include ly phoma, myeloma, hairy-cell leukemia and some viral diseases including venereal warts and rhinoviruses.
  • Immunotoxin therapy is appropriate for diseases against which the targeted antibody is effective, usually cancer. In particular, immunotoxins are being targeted for such cancers as breast cancer.
  • the dose and dosage regimen of the immunotoxin will depend, for example, upon the pharmacokinetics of the drug, the nature of the cancer (primary or metastatic) and its population, the type and length of polymer, the characteristics of the particular immunotoxin, e.g., its therapeutic index, the patient, and the patient's history.
  • the dose and dosage " regimen of the IL-2 and IFN- ⁇ will similarly depend, for example, on the pharmacokinetics of the drug, the nature of the disease, the characteristics of the IL-2 or IFN- ⁇ , the patient and the patient's history. For example, different modified IL-2 proteins are expected to have different phar acokinetic and therapeutic properties which are advantageous for different routes of administration.
  • a long-acting drug might only be administered every 3-4 days, every week or once every two weeks.
  • the clearance rate can be varied to give ultimate flexibility to fit the particular need of the patient by changing, e.g., the type of polymer and the size of the polymer attached.
  • all parts and percentages are by weight unless otherwise noted, and all temperatures are in degrees Celsius.
  • a linear, monomethyl substituted ester of PEG of molecular weight 5000 can be obtained by first reacting monomethyl PEG-5000, which is commercially available, with glutaric anhydride at 100 to 8
  • PEG- glutarate is reacted with N-hydroxysuccinimide in the presence of dicyclohexylcarbodiimide, as described in detail by Abuchowski et al., supra, on page 176.
  • the resulting product is methoxypolyethylene glycolyl N-succinimidyl glutarate, hereinafter designated as PEG .
  • a PEG carboxylic ester -HNSA was prepared using HNSA in place of N- hydroxysuccinimide. This ester preparation is described by Bhatnagar et al., supra and by Nitecki et al., supra.
  • the PEG carboxylic ester- HNSA may be used as the activated PEG in the procedures described in this and subsequent examples.
  • 50 PEG /IL-2 and 100 PEG /IL-2 there was a smear in the high molecular weight region, which is characteristic of extensively PEGylated proteins, and there was no unmodified IL-2.
  • Purified PEGylated IL-2 (after HPLC-phenyl column) was completely soluble at pH 7 in aqueous buffer without any detergent or denaturants. The purified PEGylated IL-2 remained in solution and retained its bioactivity over the time tested (at least five months). The PEGylated IL-2 which was soluble at neutral pH without SDS had the following specific activities compared to unmodified IL-2 in the presence of 0.1% SDS:
  • NK Natural Killer; described in U.S. Patent 4,518,584) and LAK (Lymphokine-Activated Killer; described in Grimm et al., J. Exp. Med., _155_: 1823-41 (1982)) activities of 10 PEG*/IL-2 and 20 PEG*/IL-2 were identical to those of unmodified IL-2. Addition of free PEG (4K daltons) in a 20-fold molar excess over unmodified IL-2 did not affect NK or LAK activities. D. Stability of PEGylated IL-2 as a function of pH.
  • PEGylated IL-2 from a modification reaction using 50 moles PEG per mole of IL-2 was incubated at various pH's at room temperature for three hours and then analyzed by 14% SDS-PAGE for hydrolysis of the amide-linked PEG from the IL-2 polypeptide.
  • PEGylated IL-2 was three-fold diluted in 10% acetonitrile containing 0.1% trifluoroacetic acid (pH 2.5), and also incubated at pH 7.5, 10 and 11. Alkaline pH's were attained by the addition of NaOH to sodium borate buffer at pH 9. No evidence of hydrolysis was obtained below pH 11 under these conditions. However, at pH 11, the PEGylated IL-2 was susceptible to hydrolysis.
  • Sample A was rendered free from aggregated material by injection in D5W containing 0.1% SDS final concentration.
  • the PEGylated IL-2 samples (B and C) contained no SDS because these are completely soluble without aggregation under normal aqueous conditions.
  • Each mouse from three groups of twelve female Balb/C mice was injected with one of the three samples into the tail vein and all were bled retro-orbital ly at 1.5 min.
  • 100 ⁇ l blood samples were removed retro-orbital ly into heparinized capillary tubes.
  • Plasma was prepared immediately by centrifugation (1 min.) and an aliquot was diluted into assay medium for bioassay as described in Example I.C.
  • Figure 3 shows the pharmacokinetics of unmodified IL-2 and two preparations of PEGylated IL-2 samples.
  • the half-lives of the initial distribution of IL-2 from Figure 3 are the following:
  • PEGylated I L-2 (50 PEG*/IL-2) 35 min.
  • PEGylation of IL-2 results in a five-fold increase in circulating half-life in mice using 10 PEG /I -2, as measured by cell proliferation assays, and a more dramatic 17-fold increase in circulating half-life using 50 PEG*/IL-2.
  • IL-2 reaction were injected intravenously into twelve mice per each type of IL-2, the percent recovery of bioactivity of total injected units of IL-2 at 1.5 minutes after injection is indicated below:
  • Pharmacokinetic data of unmodified and PEGylated I L-2 were obtained after subcutaneous administration in 48 mice of 12.5 ⁇ g protein in sterile water.
  • Samples used for scapular subcutaneous injection (single 100 ⁇ l bolus) in mice included unmodified IL-2, PEGylated IL-2 (from 20 moles PEG*/mole IL-2 reaction), and PEGylated IL-2 (from 50 moles PEG /mole IL-2 reaction). All three samples were at 0.125 mg/ml.
  • the unmodified I L-2 sample contained 0.1% SDS due to its insolubility in aqueous solution at neutral pH. At various time points 100 ⁇ l samples were removed retro- orbitally into heparinized tubes as previously described.
  • Figure 4 shows the pharmacokinetics of unmodified and PEGylated IL-2's after subcutaneous injection into mice. Not only was the maximum percentage of the total I L-2 bioactivity injected found in the plasma much higher for the PEGylated molecules as indicated below, but the clearance rate of PEGylated I L-2 was significantly lowered (see Figure 4). Maximum % IL-2 Bioactivity
  • PEGylated IL-2 50 PEG*/IL-2) 17.5 F. Immune response in rabbits after injections of PEGylated I L-2 and unmodified IL-2.
  • Group A were rabbits injected with unmodified post- diafiltered des-alanyl, IL-2 from the production process described hereinabove (lot LP 304).
  • Group B were rabbits injected with PEG/IL-2 prepared from lot LP 304 using 20-fold excess PEG* as described above.
  • Group C were rabbits injected with PEG/IL-2 also prepared from LP 304 using 50-fold excess PEG .
  • Each IL-2 preparation was diluted in sterile water prior to injection.
  • Female New Zealand White rabbits (weighing ⁇ 2.2 kg) were each injected intramuscularly at two sites, 0.5 ml (1-2 X 10 5 units) per site of the appropriate IL-2 or PEGylated IL-2.
  • the antigens tested against the sera were unmodified IL-2 (LP 304), PEG/IL-2 (20-fold excess PEG*) and PEG/IL-2
  • Rabbit Al had the least IL-2- specific IgGs.
  • Group B These rabbits developed no detectable IL-2-specific IgGs. All had IL-2-specific IgMs detected to 10 2 dilutions in ELISA assays. These assays were repeated using PEG/IL-2 as the antigen on the ELISA plates, with the same result.
  • Group C These rabbits had no detectable IL-2-specific IgGs. All had IL-2-specific IgMs detected up to 10 2 dilutions in ELISA assays done with PEG/IL-2 as the antigen.
  • mice were each injected subcutaneously at the back of the neck with 6 x 10 ⁇ Methacholanthene-A (Meth A) mouse fibrosarcoma tumor cell line, obtained from Sloan-Kettering as a cell suspension from ascites fluid in Balb/c mice.
  • the mice were segregated into three groups with similar numbers of large, medium, and small tumors (5-100 mm). The three groups were then injected intraperitoneally with test material.
  • Group A received 0.5 ml of PBS containing 0.01 mg/ml PEG (monomethyl 5000).
  • Group B received 0.5 ml of tissue culture medium containing 10% calf serum + 3 ⁇ g of the PEG*/IL-2 obtained using 20-fold excess of PEG* over IL-2.
  • Group C received tissue culture medium containing 10% calf serum + 3 ⁇ g of the unmodified postdiafiltered IL-2 as described above.
  • mice treated with formulated IL-2 showed 64% tumor growth inhibition. However, by day 9 the inhibition was only 40% and the tumors were growing rapidly. The group was sacrificed to restrict suffering. The mice treated with PEG /IL-2 showed 94% tumor growth inhibition both on day 8 and day 9. These mice also were sacrificed to restrict suffering.
  • the PEG-350 IL-2 was active as tested by the IL-2 cell proliferation bioassay in vitro mentioned abo.ve in Example I.C.
  • T*hese units are units in mouse (BRMP units x 20-fold dilution) Parentheses indicate if there were fewer than 4 mice in the group.
  • IL-2 derivatives of linear dihydroxy (unsubstituted) PEG of molecular weight 400 and 1000 were prepared generally by the method described in Example III using unsubstituted PEG-400 and PEG-1000, respectively.
  • IL-2 derivative of linear, monomethyl -substituted PEG of molecular weight 10,000 was obtained generally following the method described in Example I.A. using PEG 10,000 from Union Carbide.
  • IL-2 derivatives of dihydroxy PEG of molecular weights 20,000 and 35,000 were obtained following a method similar to Abuchowski et al., supra, referred to in Example I.A. (using base in a solvent at room temperature rather than an elevated temperature) using PEG-20,000 and PEG-35,000 from Fluka.
  • the resulting modified IL-2 proteins were bioactive as assayed by the cell proliferation assay described above.
  • CPE cytopathic effect
  • a soluble recombinant ricin A which requires no solubilization to be subjected to purification and to display cytotoxicity was prepared in accordance with the procedure described as follows.
  • the coding sequence for ricin A was placed into direct reading frame with the DNA encoding leader sequence of phoA to form a putative fusion peptide, so that the leader sequence is the N- terminal portion of a leader/ricin A chimera, the ricin A sequences so disposed result in the soluble cytotoxic material.
  • the essential component is the terminated phoA leader sequence upstream of, proximal to, and out of frame with the ricin A encoding sequence, wherein the ricin A encoding sequence is initiated by an ATG codon.
  • the two coding sequences must be, of course, provided with a compatible bacterial promoter, which was the phoA promoter already associated with the leader. Additionally, production was improved in the presence of a positive retroregulator sequence which was the positive retroregulator sequences associated with the crystal protein of B. thuringiensis. This was provided on bacterial transport vectors which included replicons and selectable markers.
  • the vectors were then used to transform a suitable procaryotic host, which was grown under conditions suitable for the particular host chosen, most frequently under conditions whereby the promoter placed in control of the expression system was suppressed.
  • the production of the ricin A was then induced by providing conditions which effect expression under control of the chosen promoter and the production permitted to proceed for sufficient time to effect a desired accumulation of product.
  • the protein product was then isolated by disrupting the cells and the cellular debris was removed.
  • the ricin A produced was then further purified using standard techniques known in the art as applied to freely soluble proteins. However, the efficiency of the extraction and purification was enhanced by treating partially clarified extract with phenyl sepharose.
  • the solubility of the ricin A in the sonicate was shown by its ability to remain in the supernatant when the sonicate was subjected to centrifugation at high speed, 100,000 x g for 30 minutes, to spin down insoluble proteins.
  • the activated PEG was dissolved in the ricin A solution by gentle mixing. At various time points 100 ⁇ l aliquots of the reaction mixture were desalted by the following procedure on G-25 columns to remove unreacted activated PEG and stop the PEGylation reaction: 100 ⁇ l PEG/ricin A applied
  • reaction mixture was maintained on ice from time 0.
  • a new sample of PEGylated ricin A was prepared for conjugation to an antibody.
  • About 40 mg of the ricin A described above was mixed with about 10 ml of Tris buffer, pH 8.5 " with 1% ⁇ - mercaptoethanol. This was concentrated to about 5 ml and desalted in an EDTA buffer on two G-25 columns to yield 6.37 ml total eluate.
  • a total of 24.6 mg of N-hydroxysuccinimide ester of PEG (about 2000) was added to the ricin A and the mixture was allowed to react for 15 minutes on ice.
  • the PEGylated ricin A was desalted over three G-25 columns, to yield a final eluate volume of 9.70 ml. ⁇ -Mercaptoethanol was added to 5 mM (about 0.05%). The PEGylated ricin A mixture was stored at 4°C.
  • a total of 50 ⁇ g of PEGylated ricin A mixture was injected into a preparative Zorbax GF-250 sizing column (DuPont) at a flow rate of 1 ml /min. using a buffer of 50 mM ( H 4 ) 2 S04, 40 mM NaP0 4 , pH 6.5.
  • the first peak was PEGylated ricin A, as determined by a 13.5% Mi ⁇ igel run of 5 ml fractions from a preparative fractionation.
  • the pool obtained in the first run was concentrated to about 2 ml and then purified by HPLC on a Zorbax GF-250 column by monitoring the absorbance at 280 nm. Fractions 15-23 of each run were pooled as PEGylated ricin A. The molecular weight of the PEGylated ricin A was then determined by running a molecular weight standard (BioRad) on HPLC, under conditions similar to the purification method described above. A linear regression was performed, indicating that the PEGylated ricin A had apparent molecular weights of about 22 K (l-mer), 44 K (2-mer), and 59 K (higher mers).
  • a breast monoclonal antibody designated 520C9 was deposited as Accession No. HB8696 on January 8, 1985 in the American Type Culture Collection, Rockv ' ille, MD. This antibody was reacted with 5,5'-dithiobis-(2-nitrobenzoic acid) at room temperature and then chilled, and then sufficient 2-iminothiolane (IT) was added to give 2.5 IT molecules per antibody molecule.
  • a total of 166 ⁇ l of propylene glycol was added to 0.84 ml of the IT-derivatized antibody.
  • the 2.32 ml of PEGylated ricin A chain described above was added to initiate the conjugation reaction. The mixture was incubated at room temperature for two hours.
  • the conjugation reaction mixture was applied to a Zorbax-GF- 250 sizing (gel filtration) HPLC column using an eluting buffer of 0.15 M NaP0 4 , pH 8.0. A total of 78% recovery of the purified immunocon jugate was obtained from the column.
  • the conjugate pool was filter-sterilized under a sterile hood into a sterile tube from which aliquots were sterilely pipetted into various sterile microfuge tubes.
  • a 100 ⁇ l aliquot of final sterifiltered immunotoxin was incubated with purified extracts from rabbit reticulocytes, ⁇ ⁇ - methionine, mRNA and other factors for mRNA synthesis.
  • the assay was measured without and with an inhibitor of ribosomal translation.
  • the dose response curve measures protein synthesis.
  • Control samples included a conjugate of non-PEGylated immunotoxin, ricin A chain, and PEGylated ricin A chain. The results were as follows: ID50 (M based on Test Material Ricin A Chain)
  • test breast cancer cells from cell line SKBR3-publicly available
  • 1 ml medium Forty thousand test breast cancer cells (from cell line SKBR3-publicly available) in 1 ml medium were added to a set of 8 ml glass vials, followed by the addition of PEGylated and non-PEGylated conjugate dilutions (in phosphate buffered saline (PBS) containing 100 ⁇ g/ml bovine serum albumin (BSA)). After incubation at 37°C for 22 hours, the medium was aspirated, the monolayers were washed with PBS, and methionine-free medium supplemented with 5$ methionine was added.
  • PBS phosphate buffered saline
  • BSA bovine serum albumin
  • the vials were further incubated for two hours at 37°C, the medium was removed, and the cells were washed twice with 2 ml of 10% trichloroacetic acid containing 1 mg/ml methionine. The cells were dried, scintillation fluid was added, and the radioactivity was counted in a scintillation counter. Cytotoxicity was expressed as the tissue culture inhibitory dose of conjugate that resulted in 50% of control (untreated) protein synthesis (TCID 50%). The results of the assay were the following:
  • the rate-limiting step does not appear to involve ricin A chain activity, but rather some other event, quite possibly conjugate binding, translocation, and/or intra-cellular routing.
  • Polyoxyethylated glycerol (POG) of molecular weight 5000 was custom synthesized by Polysciences. To 10 g of POG was added 2.28 g glutaric anhydride (a ten-fold excess over POG). The mixture was stirred for two hours at 110°C and cooled. This was dissolved in 20 ml CHC13 and poured slowly into 500 ml ether with vigorous stirring. The product was collected and rinsed with ether to yield about 90% POG-glutarate product. This product was reacted with N- hydroxysuccinimide as described in Example I.A. to yield the active ester POG-glutaryl N-hydroxysuccinimide (POG*). Then 20 ml of 0.25 mg/ml IL-2 in 0.1 M sodium borate, pH 9, 0.1% SDS as described in Example I.B. was reacted with 5 mg of the POG at room temperature for 30 minutes.
  • the plasmids with sequences used to prepare ricin A chain and the hybridoma which produces antibody 520C9 were deposited in the American Type Culture Collection (ATCC), 12301 Parklawn Drive,
  • the present invention is seen to provide a pharmaceutical composition wherein a biologically active specific protein selectively conjugated to a PEG homopolymer or a polyoxyethylated polyol and thereby made soluble or more soluble in an aqueous medium at physiological pH is dissolved in such medium.
  • the conjugation serves not only to solubilize the normally hydrophobic water-insoluble protein in water at pH 6-8, but also increases its physiological half-life and may decrease its immunogenicity by decreasing or eliminating aggregation of the normally hydrophobic protein or by shielding its antigenic determinants.
  • the protein must be solubilized by addition of solubilizing agents such as detergents or denaturants, by raising the pH in combination with addition of a stabilizer or by lowering the pH.
  • solubilizing agents such as detergents or denaturants
  • the foregoing written specification is considered to be sufficient to enable one skilled in the art to practice the invention.
  • the present invention is not to be limited in scope by the plasmids and cell line deposited, since the deposited embodiments are intended as a single illustration of one aspect of the invention and any plasmids and cell lines which are functionally equivalent are within the scope of this invention.
  • the deposit of materials herein does not constitute an admission that the written description herein contained is inadequate to enable the practice of any aspect of the invention, including the best mode thereof, nor • are they to be construed as limiting the scope of the claims to the specific illustrations which they represent.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Epidemiology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Organic Chemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Zoology (AREA)
  • Genetics & Genomics (AREA)
  • Molecular Biology (AREA)
  • Immunology (AREA)
  • Biochemistry (AREA)
  • Toxicology (AREA)
  • Wood Science & Technology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Biophysics (AREA)
  • Analytical Chemistry (AREA)
  • Botany (AREA)
  • Biomedical Technology (AREA)
  • Biotechnology (AREA)
  • Microbiology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Engineering & Computer Science (AREA)
  • Inorganic Chemistry (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Medicinal Preparation (AREA)
  • Peptides Or Proteins (AREA)
PCT/US1986/001252 1985-06-26 1986-06-06 Solubilization of proteins for pharmaceutical compositions using polymer conjugation WO1987000056A1 (en)

Priority Applications (7)

Application Number Priority Date Filing Date Title
AT86903990T ATE59301T1 (de) 1985-06-26 1986-06-06 Solubilisierung von proteinen fuer pharmazeutische zusammensetzungen mittels polymerkonjugierung.
AU59700/86A AU580431B2 (en) 1985-06-26 1986-06-06 Solubilization of proteins for pharmaceutical compositions using polymer conjugation
DE8686903990T DE3676670D1 (de) 1985-06-26 1986-06-06 Solubilisierung von proteinen fuer pharmazeutische zusammensetzungen mittels polymerkonjugierung.
IN464/CAL/86A IN163200B (US20100223739A1-20100909-C00025.png) 1985-06-26 1986-06-23
DK097987A DK169874B1 (da) 1985-06-26 1987-02-25 Farmaceutisk sammensætning indeholdende et vandopløseligt konjugat af protein og polymer og fremgangsmåde til fremstilling af dette konjugat
NO870779A NO171295C (no) 1985-06-26 1987-02-25 Fremgangsmaate for fremstilling av et farmasoeytisk preparat i form av et biologisk aktivt konjugert protein
FI870809A FI93424C (fi) 1985-06-26 1987-02-25 Menetelmä farmaseuttisessa valmisteessa käytettävän, veteen liukenevan, biologisesti aktiivisen, konjugoidun proteiinin valmistamiseksi

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US74995585A 1985-06-26 1985-06-26
US749,955 1985-06-26

Publications (1)

Publication Number Publication Date
WO1987000056A1 true WO1987000056A1 (en) 1987-01-15

Family

ID=25015921

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1986/001252 WO1987000056A1 (en) 1985-06-26 1986-06-06 Solubilization of proteins for pharmaceutical compositions using polymer conjugation

Country Status (17)

Country Link
EP (1) EP0229108B1 (US20100223739A1-20100909-C00025.png)
JP (1) JP2524586B2 (US20100223739A1-20100909-C00025.png)
KR (1) KR900004801B1 (US20100223739A1-20100909-C00025.png)
CA (1) CA1291708C (US20100223739A1-20100909-C00025.png)
DE (1) DE3676670D1 (US20100223739A1-20100909-C00025.png)
DK (1) DK169874B1 (US20100223739A1-20100909-C00025.png)
FI (1) FI93424C (US20100223739A1-20100909-C00025.png)
GR (1) GR861641B (US20100223739A1-20100909-C00025.png)
IE (1) IE59406B1 (US20100223739A1-20100909-C00025.png)
IL (1) IL79235A (US20100223739A1-20100909-C00025.png)
IN (1) IN163200B (US20100223739A1-20100909-C00025.png)
MX (1) MX174442B (US20100223739A1-20100909-C00025.png)
NZ (1) NZ216618A (US20100223739A1-20100909-C00025.png)
PH (1) PH25004A (US20100223739A1-20100909-C00025.png)
PT (1) PT82834B (US20100223739A1-20100909-C00025.png)
WO (1) WO1987000056A1 (US20100223739A1-20100909-C00025.png)
ZA (1) ZA864766B (US20100223739A1-20100909-C00025.png)

Cited By (117)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0247860A2 (en) * 1986-05-29 1987-12-02 Cetus Oncology Corporation Tumor necrosis factor formulation and its preparation
US4745180A (en) * 1986-06-27 1988-05-17 Cetus Corporation Solubilization of proteins for pharmaceutical compositions using heparin fragments
WO1989005158A1 (en) * 1987-12-10 1989-06-15 Cetus Corporation Low ph pharmaceutical compositions of recombinant beta-interferon
US4847325A (en) * 1988-01-20 1989-07-11 Cetus Corporation Conjugation of polymer to colony stimulating factor-1
EP0350230A2 (en) * 1988-07-07 1990-01-10 RESEARCH DEVELOPMENT FOUNDATION (a Nevada corporation) Immunoconjugates for cancer diagnosis and therapy
WO1990007938A1 (en) * 1989-01-23 1990-07-26 Cetus Corporation Preparation of a polymer/interleukin-2 conjugate
WO1990012874A2 (en) * 1989-04-21 1990-11-01 Genetics Institute, Inc. Cysteine added variants of polypeptides and chemical modifications thereof
WO1990015628A1 (en) * 1989-06-14 1990-12-27 Cetus Corporation Polymer/antibiotic conjugate
WO1991001758A1 (en) * 1989-08-07 1991-02-21 Debiopharm S.A. Biologically active drug polymer derivatives
EP0442724A2 (en) * 1990-02-13 1991-08-21 Kirin-Amgen, Inc. Modified hIL-6
WO1991012263A1 (fr) * 1990-02-19 1991-08-22 Serbio Bipeptides symetriques comportant un reste polyalkyleneglycol, procede de preparation et utilisation dans le dosage des proteases
WO1991012338A1 (fr) * 1990-02-19 1991-08-22 Serbio SUBSTRATS PEPTIDIQUES POUR IDENTIFICATION DU FACTEUR Xa
GB2246295A (en) * 1990-07-23 1992-01-29 Ici Plc Continuous release pharmaceutical compositions
US5091176A (en) * 1988-11-02 1992-02-25 W. R. Grace & Co.-Conn. Polymer-modified peptide drugs having enhanced biological and pharmacological activities
US5206344A (en) * 1985-06-26 1993-04-27 Cetus Oncology Corporation Interleukin-2 muteins and polymer conjugation thereof
US5229366A (en) * 1990-10-23 1993-07-20 Fuji Photo Film Co., Ltd. Peptide-containing polyethylene glycol derivatives and application thereof
US5312903A (en) * 1990-06-01 1994-05-17 E. I. Du Pont De Nemours And Company Lysine-glycosylated recombinant interleukin-2
US5334379A (en) * 1989-07-14 1994-08-02 American Cyanamid Company Cytokine and hormone carriers for conjugate vaccines
US5382657A (en) * 1992-08-26 1995-01-17 Hoffmann-La Roche Inc. Peg-interferon conjugates
US5393735A (en) * 1990-08-09 1995-02-28 Rohm And Haas Company Herbicidal glutarimides
EP0642798A2 (en) * 1993-09-08 1995-03-15 La Jolla Pharmaceutical Company Chemically-defined non-polymeric valency platform molecules and conjugates thereof
US5446090A (en) * 1993-11-12 1995-08-29 Shearwater Polymers, Inc. Isolatable, water soluble, and hydrolytically stable active sulfones of poly(ethylene glycol) and related polymers for modification of surfaces and molecules
US5539063A (en) * 1991-03-25 1996-07-23 Hoffmann-La Roche Inc. Polymer for making poly(ethylene glycol)-protein conjugates
EP0722318A1 (en) * 1993-09-08 1996-07-24 La Jolla Pharmaceutical Chemically-defined non-polymeric valency platform molecules and conjugates thereof
US5629384A (en) * 1994-05-17 1997-05-13 Consiglio Nazionale Delle Ricerche Polymers of N-acryloylmorpholine activated at one end and conjugates with bioactive materials and surfaces
EP0807115A1 (en) * 1995-01-30 1997-11-19 Enzon, Inc. High molecular weight polymer-based prodrugs
US5824778A (en) * 1988-12-22 1998-10-20 Kirin-Amgen, Inc. Chemically-modified G-CSF
WO1999055377A2 (en) * 1998-04-28 1999-11-04 Applied Research Systems Ars Holding N.V. Polyol-ifn-beta conjugates
US6004579A (en) * 1995-09-14 1999-12-21 Lxr Biotechnology, Inc. Compositions which inhibit apoptosis, methods of making the compositions and uses thereof
WO2000023114A2 (en) * 1998-10-16 2000-04-27 Biogen, Inc. Polymer conjugates of interferon beta- 1a and their uses
US6166183A (en) * 1992-11-30 2000-12-26 Kirin-Amgen, Inc. Chemically-modified G-CSF
US6207160B1 (en) 1995-06-07 2001-03-27 La Jolla Pharmaceutical Company aPL immunoreactive peptides, conjugates thereof and methods of treatment for aPL antibody-mediated pathologies
TR200101086A3 (US20100223739A1-20100909-C00025.png) * 1999-10-15 2001-08-21
US6321909B1 (en) 1997-02-13 2001-11-27 Sky High, Llc System for storing polyethylene glycol solutions
US6399578B1 (en) 1998-12-09 2002-06-04 La Jolla Pharmaceutical Company Conjugates comprising galactose α1,3 galactosyl epitopes and methods of using same
US6458953B1 (en) 1998-12-09 2002-10-01 La Jolla Pharmaceutical Company Valency platform molecules comprising carbamate linkages
US6541620B1 (en) 1989-07-18 2003-04-01 Angen Inc. Nucleic acids encoding TNF inhibitor and method of production
US6552170B1 (en) 1990-04-06 2003-04-22 Amgen Inc. PEGylation reagents and compounds formed therewith
US6800735B2 (en) 1998-10-16 2004-10-05 Biogen, Inc. Interferon-beta fusion proteins and uses
US6858210B1 (en) 1998-06-09 2005-02-22 La Jolla Pharmaceutical Co. Therapeutic and diagnostic domain 1 β2GPI polypeptides and methods of using same
WO2005019260A1 (ja) * 2003-08-25 2005-03-03 Toray Industries, Inc. インターフェロンβ複合体
US6989147B2 (en) 1996-07-09 2006-01-24 Amgen Inc. Truncated soluble tumor necrosis factor type-I and type-II receptors
US7083957B2 (en) 2001-02-12 2006-08-01 Reasearch Development Foundation Modified proteins, designer toxins, and methods of making thereof
US7090835B2 (en) 1994-10-12 2006-08-15 Amgen, Inc. N-terminally chemically modified protein compositions and methods
US7101977B2 (en) 2001-07-17 2006-09-05 Research Development Foundation Therapeutic agents comprising pro-apoptotic proteins
US7125843B2 (en) 2001-10-19 2006-10-24 Neose Technologies, Inc. Glycoconjugates including more than one peptide
US7138371B2 (en) 2001-10-10 2006-11-21 Neose Technologies, Inc Remodeling and glycoconjugation of peptides
US7157277B2 (en) 2001-11-28 2007-01-02 Neose Technologies, Inc. Factor VIII remodeling and glycoconjugation of Factor VIII
US7173003B2 (en) 2001-10-10 2007-02-06 Neose Technologies, Inc. Granulocyte colony stimulating factor: remodeling and glycoconjugation of G-CSF
US7179617B2 (en) 2001-10-10 2007-02-20 Neose Technologies, Inc. Factor IX: remolding and glycoconjugation of Factor IX
US7201897B2 (en) 1996-05-31 2007-04-10 Hoffmann-La Roche Inc. Interferon conjugates
US7214660B2 (en) 2001-10-10 2007-05-08 Neose Technologies, Inc. Erythropoietin: remodeling and glycoconjugation of erythropoietin
US7226903B2 (en) 2001-10-10 2007-06-05 Neose Technologies, Inc. Interferon beta: remodeling and glycoconjugation of interferon beta
US7265084B2 (en) 2001-10-10 2007-09-04 Neose Technologies, Inc. Glycopegylation methods and proteins/peptides produced by the methods
US7265085B2 (en) 2001-10-10 2007-09-04 Neose Technologies, Inc. Glycoconjugation methods and proteins/peptides produced by the methods
US7297511B2 (en) 2001-10-10 2007-11-20 Neose Technologies, Inc. Interferon alpha: remodeling and glycoconjugation of interferon alpha
EP1882744A1 (en) * 2006-03-27 2008-01-30 Smirnov, Mikhail Nikolaevich Method for producing an interleukin-2 preparation and the thus obtainable preparation
US7371371B2 (en) 2001-08-13 2008-05-13 University Of Southern California Interleukin-2 mutants with reduced toxicity
EP1939216A1 (en) * 2006-03-27 2008-07-02 Smirnov, Mikhail Nikolaevich Immunomodulating composition
US7399613B2 (en) 2001-10-10 2008-07-15 Neose Technologies, Inc. Sialic acid nucleotide sugars
US7405198B2 (en) 2003-11-24 2008-07-29 Neose Technologies, Inc. Glycopegylated erythropoietin
US7439043B2 (en) 2001-10-10 2008-10-21 Neose Technologies, Inc. Galactosyl nucleotide sugars
US7473680B2 (en) 2001-11-28 2009-01-06 Neose Technologies, Inc. Remodeling and glycoconjugation of peptides
EP2055189A1 (en) 2003-04-09 2009-05-06 Neose Technologies, Inc. Glycopegylation methods and proteins/peptides produced by the methods
DE202008017456U1 (de) 2007-08-27 2009-08-27 Biogenerix Ag Flüssig-Formulierung von G-CSF-Konjugaten
US7691603B2 (en) 2003-04-09 2010-04-06 Novo Nordisk A/S Intracellular formation of peptide conjugates
US7696163B2 (en) 2001-10-10 2010-04-13 Novo Nordisk A/S Erythropoietin: remodeling and glycoconjugation of erythropoietin
EP2196538A1 (en) * 2000-02-10 2010-06-16 Mountain View Pharmaceuticals, Inc. Conjugates prepared with aggregate-free proteins
US7785579B2 (en) * 1997-01-15 2010-08-31 Polaris Group Modified tumor necrosis factor
US7795210B2 (en) 2001-10-10 2010-09-14 Novo Nordisk A/S Protein remodeling methods and proteins/peptides produced by the methods
US7803777B2 (en) 2003-03-14 2010-09-28 Biogenerix Ag Branched water-soluble polymers and their conjugates
EP2298354A2 (en) 2001-10-10 2011-03-23 BioGeneriX AG Remodelling and glycoconjugation of interferon-beta
US7927589B2 (en) 1998-08-06 2011-04-19 Mountain View Pharmaceuticals, Inc. PEG-urate oxidase conjugates and use thereof
US8017733B2 (en) 2002-01-18 2011-09-13 Biogen Idec Ma Inc. Polyalkylene polymer compounds and uses thereof
EP2386571A2 (en) 2005-04-08 2011-11-16 BioGeneriX AG Compositions and methods for the preparation of protease resistant human growth hormone glycosylation mutants
US8071737B2 (en) 1995-05-04 2011-12-06 Glead Sciences, Inc. Nucleic acid ligand complexes
CN102319437A (zh) * 2002-12-26 2012-01-18 山景医药公司 具有增强的生物学效用的干扰素-β的聚合物缀合物
WO2012107417A1 (en) 2011-02-10 2012-08-16 Roche Glycart Ag Mutant interleukin-2 polypeptides
US8268967B2 (en) 2004-09-10 2012-09-18 Novo Nordisk A/S Glycopegylated interferon α
EP2514757A2 (en) 2005-01-10 2012-10-24 BioGeneriX AG Glycopegylated granulocyte colony stimulating factor
WO2013129549A1 (ja) 2012-02-29 2013-09-06 東レ株式会社 体腔液貯留抑制剤
WO2013156488A2 (en) 2012-04-16 2013-10-24 Leverton Licence Holdings Limited Optimised subcutaneous therapeutic agents
EP2672259A1 (en) 2008-05-13 2013-12-11 Advanced Liquid Logic, Inc. Droplet actuator devices, systems and methods
WO2013177187A3 (en) * 2012-05-22 2014-01-16 Massachusetts Institute Of Technology Synergistic tumor treatment with extended-pk il-2 and therapeutic agents
US8791066B2 (en) 2004-07-13 2014-07-29 Novo Nordisk A/S Branched PEG remodeling and glycosylation of glucagon-like peptide-1 [GLP-1]
US8791070B2 (en) 2003-04-09 2014-07-29 Novo Nordisk A/S Glycopegylated factor IX
US8841439B2 (en) 2005-11-03 2014-09-23 Novo Nordisk A/S Nucleotide sugar purification using membranes
US8911967B2 (en) 2005-08-19 2014-12-16 Novo Nordisk A/S One pot desialylation and glycopegylation of therapeutic peptides
US8916360B2 (en) 2003-11-24 2014-12-23 Novo Nordisk A/S Glycopegylated erythropoietin
US20150017120A1 (en) * 2013-06-13 2015-01-15 Massachusetts Institute Of Technology Synergistic tumor treatment with extended-pk il-2 and adoptive cell therapy
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
US9005625B2 (en) 2003-07-25 2015-04-14 Novo Nordisk A/S Antibody toxin conjugates
US9023992B2 (en) 2004-05-04 2015-05-05 Novo Nordisk Healthcare Ag Hydrophobic interaction chromatography purification of factor VII polypeptides
US9050304B2 (en) 2007-04-03 2015-06-09 Ratiopharm Gmbh Methods of treatment using glycopegylated G-CSF
US9150848B2 (en) 2008-02-27 2015-10-06 Novo Nordisk A/S Conjugated factor VIII molecules
EP2933264A2 (en) 2008-01-22 2015-10-21 Araim Pharmaceuticals, Inc. Tissue protective peptides and peptide analogs for preventing and treating diseases and disorders associated with tissue damage
US9187532B2 (en) 2006-07-21 2015-11-17 Novo Nordisk A/S Glycosylation of peptides via O-linked glycosylation sequences
US9200049B2 (en) 2004-10-29 2015-12-01 Novo Nordisk A/S Remodeling and glycopegylation of fibroblast growth factor (FGF)
EP2975135A1 (en) 2005-05-25 2016-01-20 Novo Nordisk A/S Glycopegylated factor IX
US9493499B2 (en) 2007-06-12 2016-11-15 Novo Nordisk A/S Process for the production of purified cytidinemonophosphate-sialic acid-polyalkylene oxide (CMP-SA-PEG) as modified nucleotide sugars via anion exchange chromatography
WO2018184964A1 (en) 2017-04-03 2018-10-11 F. Hoffmann-La Roche Ag Immunoconjugates of an anti-pd-1 antibody with a mutant il-2 or with il-15
WO2018184965A1 (en) 2017-04-03 2018-10-11 F. Hoffmann-La Roche Ag Immunoconjugates of il-2 with an anti-pd-1 and tim-3 bispecific antibody
US10139399B2 (en) 2009-06-25 2018-11-27 Horizon Pharma Rheumatology Llc Methods and kits for predicting infusion reaction risk and antibody-mediated loss of response by monitoring serum uric acid during PEGylated uricase therapy
US10160958B2 (en) 2005-04-11 2018-12-25 Horizon Pharma Rheumatology Llc Variant forms of urate oxidase and use thereof
WO2020158690A1 (ja) 2019-01-28 2020-08-06 東レ株式会社 肝細胞増殖因子又はその活性断片のポリエチレングリコール修飾体
WO2020158691A1 (ja) 2019-01-28 2020-08-06 東レ株式会社 肝細胞増殖因子又はその活性断片のポリエチレングリコール修飾体
US10864253B2 (en) 2016-04-29 2020-12-15 Araim Pharmaceuticals, Inc. Tissue protective peptides for preventing and treating diseases and disorders associated with tissue damage
WO2021001289A1 (en) 2019-07-02 2021-01-07 F. Hoffmann-La Roche Ag Immunoconjugates comprising a mutant interleukin-2 and an anti-cd8 antibody
US20210023230A1 (en) * 2010-11-12 2021-01-28 Nektar Therapeutics Conjugates of an il-2 moiety and a polymer
WO2021209402A2 (en) 2020-04-15 2021-10-21 F. Hoffmann-La Roche Ag Immunoconjugates
WO2021231278A1 (en) 2020-05-11 2021-11-18 F. Hoffmann-La Roche Ag Combination therapy with modified pbmcs and an immunoconjugate
WO2022117692A2 (en) 2020-12-04 2022-06-09 F. Hoffmann-La Roche Ag Ph-dependent mutant interleukin-2 polypeptides
WO2022148853A1 (en) 2021-01-11 2022-07-14 F. Hoffmann-La Roche Ag Immunoconjugates
WO2023052541A1 (en) 2021-09-30 2023-04-06 Imcheck Therapeutics Combination of an anti-btn3a activating antibody and an il-2 agonist for use in therapy
WO2023062050A1 (en) 2021-10-14 2023-04-20 F. Hoffmann-La Roche Ag New interleukin-7 immunoconjugates
WO2023062048A1 (en) 2021-10-14 2023-04-20 F. Hoffmann-La Roche Ag Alternative pd1-il7v immunoconjugates for the treatment of cancer
AU2018394189B2 (en) * 2017-12-27 2023-12-21 Kyowa Kirin Co., Ltd. IL-2 variant

Families Citing this family (55)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030053982A1 (en) 1994-09-26 2003-03-20 Kinstler Olaf B. N-terminally chemically modified protein compositions and methods
ES2336857T3 (es) * 1995-05-04 2010-04-16 Gilead Sciences, Inc. Complejos de ligandos de acido nucleico.
US7144574B2 (en) 1999-08-27 2006-12-05 Maxygen Aps Interferon β variants and conjugates
US7431921B2 (en) 2000-04-14 2008-10-07 Maxygen Aps Interferon beta-like molecules
HUP0302674A2 (hu) * 1999-08-27 2003-11-28 Maxigen Aps. Új, béta-interferon-szerű molekulák
US6531122B1 (en) 1999-08-27 2003-03-11 Maxygen Aps Interferon-β variants and conjugates
EP1231943A1 (en) 1999-11-12 2002-08-21 Maxygen Holdings Ltd Interferon gamma conjugates
US7230081B1 (en) 1999-11-12 2007-06-12 Maxygen Holdings, Ltd. Interferon gamma conjugates
ATE435033T1 (de) 2000-01-10 2009-07-15 Maxygen Holdings Ltd G-csf konjugate
EP2319541A1 (en) 2000-02-11 2011-05-11 Bayer HealthCare LLC Factor VII or VIIA-like conjugates
EP1366075B1 (en) 2001-02-27 2009-05-27 Maxygen Aps New interferon beta-like molecules
US7038015B2 (en) 2001-04-06 2006-05-02 Maxygen Holdings, Ltd. Interferon gamma polypeptide variants
US6958388B2 (en) 2001-04-06 2005-10-25 Maxygen, Aps Interferon gamma polypeptide variants
WO2003000278A1 (fr) * 2001-06-22 2003-01-03 Kyowa Hakko Kogyo Co., Ltd. Onguents
UA86744C2 (en) 2002-06-21 2009-05-25 Ново Нордиск Хэлс Кеа Аг Pegylated factor vii glycoforms
US7524931B2 (en) 2002-07-03 2009-04-28 Maxygen Holdings Ltd. Full-length interferon gamma polypeptide variants
WO2005023862A2 (en) * 2003-08-07 2005-03-17 Zymogenetics, Inc. Homogeneous preparations of il-28 and il-29
CN102516386A (zh) 2003-10-10 2012-06-27 诺沃挪第克公司 Il-21衍生物
EP2633866A3 (en) 2003-10-17 2013-12-18 Novo Nordisk A/S Combination therapy
BRPI0507169A (pt) 2004-02-02 2007-06-26 Ambrx Inc polipeptìdeos do hormÈnio de crescimento humano modificados e seu usos
KR20070034512A (ko) 2004-06-18 2007-03-28 암브룩스, 인코포레이티드 신규 항원-결합 폴리펩티드 및 이의 용도
CA2594557C (en) 2004-12-22 2016-04-26 Ambrx, Inc. Modified human growth hormone
WO2006134173A2 (en) 2005-06-17 2006-12-21 Novo Nordisk Health Care Ag Selective reduction and derivatization of engineered proteins comprising at least one non-native cysteine
CN101454461A (zh) 2005-11-16 2009-06-10 Ambrx公司 包括非天然氨基酸的方法和组合物
EP2213733A3 (en) 2006-05-24 2010-12-29 Novo Nordisk Health Care AG Factor IX analogues having prolonged in vivo half life
CN106008699A (zh) 2006-09-08 2016-10-12 Ambrx公司 经修饰的人类血浆多肽或Fc骨架和其用途
MX2009002820A (es) * 2006-09-15 2009-06-04 Creabilis Therapeutics Spa Conjugados polimericos de box-a de hmgb1 y variantes de box-a de hmgb1.
KR101476472B1 (ko) 2007-03-30 2015-01-05 암브룩스, 인코포레이티드 변형된 fgf-21 폴리펩티드 및 그 용도
ES2632504T3 (es) 2007-11-20 2017-09-13 Ambrx, Inc. Polipéptidos de insulina modificados y sus usos
JP2011510005A (ja) * 2008-01-18 2011-03-31 ダウ グローバル テクノロジーズ インコーポレイティド メトキシポリエチレングリコールを用いる難溶性活性物質の水溶解度増強方法
NZ602170A (en) 2008-02-08 2014-03-28 Ambrx Inc Modified leptin polypeptides and their uses
JP5680534B2 (ja) 2008-07-23 2015-03-04 イーライ リリー アンド カンパニー 修飾されているウシg−csfポリペプチドおよびそれらの使用
MX2011003272A (es) 2008-09-26 2011-04-28 Ambrx Inc Polipeptidos de eritropoyetina animal modificados y sus usos.
ES2660000T3 (es) 2008-09-26 2018-03-20 Ambrx, Inc. Vacunas y microorganismos dependientes de la replicación de aminoácidos no naturales
JP2013515080A (ja) 2009-12-21 2013-05-02 アンブルックス,インコーポレイテッド 修飾されているウシのソマトトロピンポリペプチドおよびそれらの使用
EP2805965A1 (en) 2009-12-21 2014-11-26 Ambrx, Inc. Modified porcine somatotropin polypeptides and their uses
EP2542569B1 (en) 2010-03-05 2020-09-16 Omeros Corporation Chimeric inhibitor molecules of complement activation
EP2569331A1 (en) 2010-05-10 2013-03-20 Perseid Therapeutics LLC Polypeptide inhibitors of vla4
US9567386B2 (en) 2010-08-17 2017-02-14 Ambrx, Inc. Therapeutic uses of modified relaxin polypeptides
ES2972902T3 (es) 2010-08-17 2024-06-17 Ambrx Inc Polipéptidos de relaxina modificados y sus usos
TWI480288B (zh) 2010-09-23 2015-04-11 Lilly Co Eli 牛顆粒細胞群落刺激因子及其變體之調配物
KR20140054009A (ko) 2011-07-01 2014-05-08 바이엘 인텔렉쳐 프로퍼티 게엠베하 릴랙신 융합 폴리펩타이드 및 그의 용도
EP2859017B1 (en) 2012-06-08 2019-02-20 Sutro Biopharma, Inc. Antibodies comprising site-specific non-natural amino acid residues, methods of their preparation and methods of their use
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
SG11201501464TA (en) 2012-08-31 2015-03-30 Sutro Biopharma Inc Modified amino acids comprising an azido group
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
US9840493B2 (en) 2013-10-11 2017-12-12 Sutro Biopharma, Inc. Modified amino acids comprising tetrazine functional groups, methods of preparation, and methods of their use
AU2015335603B2 (en) 2014-10-24 2020-04-30 Bristol-Myers Squibb Company Modified FGF-21 polypeptides and uses thereof
AU2018219283B2 (en) 2017-02-08 2022-05-19 Bristol-Myers Squibb Company Modified relaxin polypeptides comprising a pharmacokinetic enhancer and uses thereof
SI3849614T1 (sl) 2018-09-11 2024-04-30 Ambrx, Inc. Polipeptidni konjugati interlevkina-2 in njihove uporabe
CN113366015A (zh) 2018-10-19 2021-09-07 Ambrx公司 白细胞介素-10多肽缀合物、其二聚体及其用途
KR20210136014A (ko) 2019-02-12 2021-11-16 암브룩스, 인코포레이티드 항체-tlr 작용제 콘쥬게이트를 함유하는 조성물, 방법 및 이의 용도
IL296099A (en) 2020-03-11 2022-11-01 Ambrx Inc Interleukin-2 polypeptide conjugates and methods of using them
US20230302150A1 (en) 2020-08-20 2023-09-28 Ambrx, Inc. Antibody-tlr agonist conjugates, methods and uses thereof
EP4313163A1 (en) 2021-04-03 2024-02-07 Ambrx, Inc. Anti-her2 antibody-drug conjugates and uses thereof

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4179337A (en) * 1973-07-20 1979-12-18 Davis Frank F Non-immunogenic polypeptides
US4414147A (en) * 1981-04-17 1983-11-08 Massachusetts Institute Of Technology Methods of decreasing the hydrophobicity of fibroblast and other interferons
EP0098110A2 (en) * 1982-06-24 1984-01-11 NIHON CHEMICAL RESEARCH KABUSHIKI KAISHA also known as JAPAN CHEMICAL RESEARCH CO., LTD Long-acting composition
EP0154316A2 (en) * 1984-03-06 1985-09-11 Takeda Chemical Industries, Ltd. Chemically modified lymphokine and production thereof

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPS54157816A (en) * 1978-05-27 1979-12-13 Unitika Ltd Fixing of bioactive substance to solid surface
WO1985003934A1 (en) * 1984-03-06 1985-09-12 Takeda Chemical Industries, Ltd. Chemically modified protein and process for its preparation

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4179337A (en) * 1973-07-20 1979-12-18 Davis Frank F Non-immunogenic polypeptides
US4414147A (en) * 1981-04-17 1983-11-08 Massachusetts Institute Of Technology Methods of decreasing the hydrophobicity of fibroblast and other interferons
EP0098110A2 (en) * 1982-06-24 1984-01-11 NIHON CHEMICAL RESEARCH KABUSHIKI KAISHA also known as JAPAN CHEMICAL RESEARCH CO., LTD Long-acting composition
EP0154316A2 (en) * 1984-03-06 1985-09-11 Takeda Chemical Industries, Ltd. Chemically modified lymphokine and production thereof

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
CHEMICAL ABSTRACTS, Volume 101, No. 15, 8 October 1984 (Columbus, Ohio, US) A. ABUCHOWSKI et al.: "Cancer Therapy with Chemically Modified Enzymes. I. Antitumor Properties of Polyethylene Glycol-Asparaginase Conjugates", see page 29, Abstract 122679c, & Cancer Biochem. Biophys. 1984, 7(2), 175-86 cited in the application *

Cited By (236)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5206344A (en) * 1985-06-26 1993-04-27 Cetus Oncology Corporation Interleukin-2 muteins and polymer conjugation thereof
EP0247860A3 (en) * 1986-05-29 1989-10-11 Cetus Corporation Tumor necrosis factor formulation and its preparation
EP0247860A2 (en) * 1986-05-29 1987-12-02 Cetus Oncology Corporation Tumor necrosis factor formulation and its preparation
US4745180A (en) * 1986-06-27 1988-05-17 Cetus Corporation Solubilization of proteins for pharmaceutical compositions using heparin fragments
WO1989009624A1 (en) * 1986-06-27 1989-10-19 Cetus Corporation Solubilization of proteins for pharmaceutical compositions
WO1989005158A1 (en) * 1987-12-10 1989-06-15 Cetus Corporation Low ph pharmaceutical compositions of recombinant beta-interferon
US4847325A (en) * 1988-01-20 1989-07-11 Cetus Corporation Conjugation of polymer to colony stimulating factor-1
EP0350230A2 (en) * 1988-07-07 1990-01-10 RESEARCH DEVELOPMENT FOUNDATION (a Nevada corporation) Immunoconjugates for cancer diagnosis and therapy
EP0350230A3 (en) * 1988-07-07 1990-12-12 Res Dev Foundation Immunoconjugates for cancer diagnosis and therapy
US5091176A (en) * 1988-11-02 1992-02-25 W. R. Grace & Co.-Conn. Polymer-modified peptide drugs having enhanced biological and pharmacological activities
US5824778A (en) * 1988-12-22 1998-10-20 Kirin-Amgen, Inc. Chemically-modified G-CSF
WO1990007938A1 (en) * 1989-01-23 1990-07-26 Cetus Corporation Preparation of a polymer/interleukin-2 conjugate
EP0668353A1 (en) * 1989-04-21 1995-08-23 Genetics Institute, Inc. Cysteine added variants of EPO and chemical modifications thereof
US5166322A (en) * 1989-04-21 1992-11-24 Genetics Institute Cysteine added variants of interleukin-3 and chemical modifications thereof
EP0668354A1 (en) * 1989-04-21 1995-08-23 Genetics Institute, Inc. Cysteine added variants of G-CSF and chemical modifications thereof
WO1990012874A3 (en) * 1989-04-21 1991-01-10 Genetics Inst Cysteine added variants of polypeptides and chemical modifications thereof
WO1990012874A2 (en) * 1989-04-21 1990-11-01 Genetics Institute, Inc. Cysteine added variants of polypeptides and chemical modifications thereof
WO1990015628A1 (en) * 1989-06-14 1990-12-27 Cetus Corporation Polymer/antibiotic conjugate
US5334379A (en) * 1989-07-14 1994-08-02 American Cyanamid Company Cytokine and hormone carriers for conjugate vaccines
US6541620B1 (en) 1989-07-18 2003-04-01 Angen Inc. Nucleic acids encoding TNF inhibitor and method of production
WO1991001758A1 (en) * 1989-08-07 1991-02-21 Debiopharm S.A. Biologically active drug polymer derivatives
EP0442724A3 (en) * 1990-02-13 1992-02-26 Kirin-Amgen, Inc. Modified hil-6
EP0442724A2 (en) * 1990-02-13 1991-08-21 Kirin-Amgen, Inc. Modified hIL-6
AU634343B2 (en) * 1990-02-13 1993-02-18 Kirin-Amgen Incorporated Modified hil-6
US5264209A (en) * 1990-02-13 1993-11-23 Kirin-Amgen, Inc. Modified HIL-6
FR2658521A1 (fr) * 1990-02-19 1991-08-23 Serbioo Bipeptides symetriques comportant un reste polyalkyleneglycol, procede de preparation et utilisation dans le dosage des proteases.
WO1991012263A1 (fr) * 1990-02-19 1991-08-22 Serbio Bipeptides symetriques comportant un reste polyalkyleneglycol, procede de preparation et utilisation dans le dosage des proteases
US5225532A (en) * 1990-02-19 1993-07-06 Serbio Symmetrical bipeptides containing a polyalkylene glycol residue, method of preparation and use in the assay of proteases
WO1991012338A1 (fr) * 1990-02-19 1991-08-22 Serbio SUBSTRATS PEPTIDIQUES POUR IDENTIFICATION DU FACTEUR Xa
FR2658519A1 (fr) * 1990-02-19 1991-08-23 Serbio Substrats peptidiques pour identification du facteur xa.
US6552170B1 (en) 1990-04-06 2003-04-22 Amgen Inc. PEGylation reagents and compounds formed therewith
US5312903A (en) * 1990-06-01 1994-05-17 E. I. Du Pont De Nemours And Company Lysine-glycosylated recombinant interleukin-2
GB2246295A (en) * 1990-07-23 1992-01-29 Ici Plc Continuous release pharmaceutical compositions
US5320840A (en) * 1990-07-23 1994-06-14 Imperial Chemical Industries Plc Continuous release pharmaceutical compositions
EP0473268A3 (en) * 1990-07-23 1992-09-16 Imperial Chemical Industries Plc Continuous release pharmaceutical compositions comprising a polypeptide covalently conjugated to a water soluble polymer
GB2246295B (en) * 1990-07-23 1994-05-11 Ici Plc Continuous release pharmaceutical compositions
US5773581A (en) * 1990-07-23 1998-06-30 Zeneca Limited Conjugate of a solution stable G-CSF derivative and a water-soluble polymer
EP0473268A2 (en) * 1990-07-23 1992-03-04 Zeneca Limited Continuous release pharmaceutical compositions comprising a polypeptide covalently conjugated to a water soluble polymer
US5393735A (en) * 1990-08-09 1995-02-28 Rohm And Haas Company Herbicidal glutarimides
US5229366A (en) * 1990-10-23 1993-07-20 Fuji Photo Film Co., Ltd. Peptide-containing polyethylene glycol derivatives and application thereof
US5595732A (en) * 1991-03-25 1997-01-21 Hoffmann-La Roche Inc. Polyethylene-protein conjugates
US5792834A (en) * 1991-03-25 1998-08-11 Hoffmann-La Roche Inc. Polyethylene-protein conjugates
US5849860A (en) * 1991-03-25 1998-12-15 Hoffmann-La Roche Inc. Polyethylene-protein conjugates
US5834594A (en) * 1991-03-25 1998-11-10 Hoffman-La Roche Inc. Polyethylene-protein conjugates
US5539063A (en) * 1991-03-25 1996-07-23 Hoffmann-La Roche Inc. Polymer for making poly(ethylene glycol)-protein conjugates
US5747646A (en) * 1991-03-25 1998-05-05 Hoffmann-La Roche Inc. Polyethylene-protein conjugates
US5559213A (en) * 1991-03-25 1996-09-24 Hoffmann-La Roche Inc. Polyethylene-protein conjugates
US5382657A (en) * 1992-08-26 1995-01-17 Hoffmann-La Roche Inc. Peg-interferon conjugates
EP0593868B1 (en) * 1992-08-26 1998-04-15 F. Hoffmann-La Roche Ag PEG-interferon conjugates
US6166183A (en) * 1992-11-30 2000-12-26 Kirin-Amgen, Inc. Chemically-modified G-CSF
EP0642798A3 (en) * 1993-09-08 1998-09-16 La Jolla Pharmaceutical Company Chemically-defined non-polymeric valency platform molecules and conjugates thereof
EP0722318A1 (en) * 1993-09-08 1996-07-24 La Jolla Pharmaceutical Chemically-defined non-polymeric valency platform molecules and conjugates thereof
EP0642798A2 (en) * 1993-09-08 1995-03-15 La Jolla Pharmaceutical Company Chemically-defined non-polymeric valency platform molecules and conjugates thereof
EP1808183A3 (en) * 1993-09-08 2009-07-01 La Jolla Pharmaceutical Company Chemically-defined non-polymeric valency platform molecules and conjugates thereof
EP1808183A2 (en) * 1993-09-08 2007-07-18 La Jolla Pharmaceutical Company Chemically-defined non-polymeric valency platform molecules and conjugates thereof
EP0722318A4 (en) * 1993-09-08 1998-08-05 Jolla Pharma NON-POLYMERIC VALIDATED CHEMICALLY DEFINED PLATFORM MOLECULES AND THEIR CONJUGATES
CN1318023C (zh) * 1993-09-08 2007-05-30 拉卓拉药物公司 化学上定义的非聚合价平台分子和其偶联物
US6894025B2 (en) 1993-11-12 2005-05-17 Nektar Therapeutics Al, Corp. Biologically active molecules having thiol moiety conjugated to polymers containing ethyl sulfone moiety
US5739208A (en) * 1993-11-12 1998-04-14 Shearwater Polymers, Inc. Isolatable, water soluble, and hydrolytically stable active sulfones of poly(ethylene glycol) and related polymers for modification of surfaces and molecules
US5446090A (en) * 1993-11-12 1995-08-29 Shearwater Polymers, Inc. Isolatable, water soluble, and hydrolytically stable active sulfones of poly(ethylene glycol) and related polymers for modification of surfaces and molecules
US5900461A (en) * 1993-11-12 1999-05-04 Shearwater Polymers, Inc. Isolatable, water soluble, and hydrolytically stable active sulfones of poly(ethylene glycol) and related polymers for modification of surfaces and molecules
US7214366B2 (en) 1993-11-12 2007-05-08 Nektar Therapeutics Al, Corporation Isolatable, water soluble, and hydrolytically stable active sulfones of poly(ethylene glycol) and related polymers for modification of surfaces and molecules
US5631322A (en) * 1994-05-17 1997-05-20 Consiglio Nazionale Delle Ricerche Polymers of N-acryloylmorpholine activated at one end and conjugates with bioactive materials and surfaces
US5629384A (en) * 1994-05-17 1997-05-13 Consiglio Nazionale Delle Ricerche Polymers of N-acryloylmorpholine activated at one end and conjugates with bioactive materials and surfaces
US7090835B2 (en) 1994-10-12 2006-08-15 Amgen, Inc. N-terminally chemically modified protein compositions and methods
US8258262B2 (en) 1994-10-12 2012-09-04 Amgen Inc. N-terminally chemically modified protein compositions and methods
US7662933B2 (en) 1994-10-12 2010-02-16 Amgen Inc. N-terminally chemically modified protein compositions and methods
EP0807115A1 (en) * 1995-01-30 1997-11-19 Enzon, Inc. High molecular weight polymer-based prodrugs
EP0807115A4 (en) * 1995-01-30 1998-10-07 Enzon Inc MEDICAMENTS BASED ON POLYMERS WITH HIGH MOLECULAR WEIGHT
US8071737B2 (en) 1995-05-04 2011-12-06 Glead Sciences, Inc. Nucleic acid ligand complexes
US6207160B1 (en) 1995-06-07 2001-03-27 La Jolla Pharmaceutical Company aPL immunoreactive peptides, conjugates thereof and methods of treatment for aPL antibody-mediated pathologies
US6004579A (en) * 1995-09-14 1999-12-21 Lxr Biotechnology, Inc. Compositions which inhibit apoptosis, methods of making the compositions and uses thereof
US7201897B2 (en) 1996-05-31 2007-04-10 Hoffmann-La Roche Inc. Interferon conjugates
US7732587B2 (en) 1996-07-09 2010-06-08 Amgen Inc. Nucleic acids encoding truncated soluble tumor necrosis factor
US6989147B2 (en) 1996-07-09 2006-01-24 Amgen Inc. Truncated soluble tumor necrosis factor type-I and type-II receptors
US7785579B2 (en) * 1997-01-15 2010-08-31 Polaris Group Modified tumor necrosis factor
US6321909B1 (en) 1997-02-13 2001-11-27 Sky High, Llc System for storing polyethylene glycol solutions
KR100622796B1 (ko) * 1998-04-28 2006-09-13 어플라이드 리서치 시스템스 에이알에스 홀딩 엔.브이. 폴리올-ifn-베타 공액체
CZ298579B6 (cs) * 1998-04-28 2007-11-14 Applied Research Systems Ars Holding N. V. Konjugát polyolu a interferonu-beta
US7700314B2 (en) 1998-04-28 2010-04-20 Merck Serono Sa Method for producing polyol-IFN-β conjugate
EA005495B1 (ru) * 1998-04-28 2005-02-24 Апплайд Резеч Системз Арс Холдинг Н.В. Способ поэтапного присоединения частей полиэтиленгликоля к полипептиду
US7357925B2 (en) 1998-04-28 2008-04-15 Laboratoires Seronosa Method for treating disorders and diseases treatable with human fibroblast interferon
AU762621B2 (en) * 1998-04-28 2003-07-03 Merck Serono Sa Polyol-IFN-beta conjugates
WO1999055377A3 (en) * 1998-04-28 1999-12-29 Applied Research Systems Polyol-ifn-beta conjugates
WO1999055377A2 (en) * 1998-04-28 1999-11-04 Applied Research Systems Ars Holding N.V. Polyol-ifn-beta conjugates
US6638500B1 (en) 1998-04-28 2003-10-28 Applied Research Systems Ars Holding N.V. Polyol-IFN-βconjugates modified at Cys-17 and composition containing same
US6858210B1 (en) 1998-06-09 2005-02-22 La Jolla Pharmaceutical Co. Therapeutic and diagnostic domain 1 β2GPI polypeptides and methods of using same
US8067553B2 (en) 1998-08-06 2011-11-29 Mountain View Pharmaceuticals, Inc. PEG-urate oxidase conjugates and use thereof
US8921064B2 (en) 1998-08-06 2014-12-30 Mountain View Pharmaceuticals, Inc. Method for purifying urate oxidase tetramers and octamers
US9885024B2 (en) 1998-08-06 2018-02-06 Duke University PEG-urate oxidase conjugates and use thereof
US8618267B2 (en) 1998-08-06 2013-12-31 Mountain View Pharmaceuticals, Inc. PEG-urate oxidase conjugates and use thereof
US7927589B2 (en) 1998-08-06 2011-04-19 Mountain View Pharmaceuticals, Inc. PEG-urate oxidase conjugates and use thereof
CN1329082C (zh) * 1998-10-16 2007-08-01 拜奥根Idec马萨诸塞公司 干扰素-β-1a的聚合物缀合物及其使用
AU762616B2 (en) * 1998-10-16 2003-07-03 Biogen Ma Inc. Polymer conjugates of interferon beta-1a and uses
WO2000023114A2 (en) * 1998-10-16 2000-04-27 Biogen, Inc. Polymer conjugates of interferon beta- 1a and their uses
US7446173B2 (en) 1998-10-16 2008-11-04 Biogen Idec Ma Inc. Polymer conjugates of interferon beta-1A and uses
US6962978B2 (en) 1998-10-16 2005-11-08 Biogen, Inc. Polymer conjugates of interferon beta-1a and uses
WO2000023114A3 (en) * 1998-10-16 2000-09-28 Biogen Inc Polymer conjugates of interferon beta- 1a and their uses
SG110047A1 (en) * 1998-10-16 2005-04-28 Biogen Inc Polymer conjugates of interferon beta-a1 and uses
CN101062419B (zh) * 1998-10-16 2016-06-29 生物基因Ma公司 干扰素-β-1a的聚合物缀合物及其使用
EP2260872A3 (en) * 1998-10-16 2011-04-27 Biogen Idec MA Inc. Polymer conjugates of interferon beta-1A and uses thereof
US6800735B2 (en) 1998-10-16 2004-10-05 Biogen, Inc. Interferon-beta fusion proteins and uses
EP2599503A3 (en) * 1998-10-16 2013-07-24 Biogen Idec MA Inc. Polymer conjugates of interferon beta-1A and uses thereof
EP2260872A2 (en) * 1998-10-16 2010-12-15 Biogen Idec MA Inc. Polymer conjugates of interferon beta-1A and uses thereof
EP1656952A3 (en) * 1998-10-16 2006-11-29 Biogen Idec MA Inc. Polymer conjugates of interferon beta-1A and uses thereof
EA004789B1 (ru) * 1998-10-16 2004-08-26 Байоджен, Инк. Полимерные конъюгаты бета-1а-интерферона и их использование
US9314534B2 (en) 1998-10-16 2016-04-19 Biogen Ma Inc. Polymer conjugates of interferon beta-1A and uses
US7527946B2 (en) 1998-10-16 2009-05-05 Biogen Idec Ma Inc., Interferon-beta-1a-immunoglobulin fusion proteins and uses
CZ299164B6 (cs) * 1998-10-16 2008-05-07 Biogen Idec Ma Inc. Farmaceutická kompozice obsahující glykosylovaný interferon-beta-1a navázaný na polymer
US6458953B1 (en) 1998-12-09 2002-10-01 La Jolla Pharmaceutical Company Valency platform molecules comprising carbamate linkages
US6399578B1 (en) 1998-12-09 2002-06-04 La Jolla Pharmaceutical Company Conjugates comprising galactose α1,3 galactosyl epitopes and methods of using same
TR200101086A3 (US20100223739A1-20100909-C00025.png) * 1999-10-15 2001-08-21
EP2196538A1 (en) * 2000-02-10 2010-06-16 Mountain View Pharmaceuticals, Inc. Conjugates prepared with aggregate-free proteins
US7741278B2 (en) 2001-02-12 2010-06-22 Research Development Foundation Modified proteins, designer toxins, and methods of making thereof
US7285635B2 (en) 2001-02-12 2007-10-23 Research Development Foundation Modified proteins, designer toxins, and methods of making thereof
US7943571B2 (en) 2001-02-12 2011-05-17 Research Development Foundation Modified proteins, designer toxins, and methods of making thereof
US8138311B2 (en) 2001-02-12 2012-03-20 Research Development Foundation Modified proteins, designer toxins, and methods of making thereof
US7083957B2 (en) 2001-02-12 2006-08-01 Reasearch Development Foundation Modified proteins, designer toxins, and methods of making thereof
US7759091B2 (en) 2001-07-17 2010-07-20 Research Development Foundation Therapeutic agents comprising pro-apoptotic proteins
US7101977B2 (en) 2001-07-17 2006-09-05 Research Development Foundation Therapeutic agents comprising pro-apoptotic proteins
US8043831B2 (en) 2001-07-17 2011-10-25 Research Development Foundation Therapeutic agents comprising pro-apoptotic proteins
US8530225B2 (en) 2001-07-17 2013-09-10 Research Development Foundation Therapeutic agents comprising pro-apoptotic proteins
US7371723B2 (en) 2001-07-17 2008-05-13 Research Development Foundation Therapeutic agents comprising pro-apoptotic proteins
US7371371B2 (en) 2001-08-13 2008-05-13 University Of Southern California Interleukin-2 mutants with reduced toxicity
US7514073B2 (en) 2001-08-13 2009-04-07 University Of Southern California Therapeutic use of interleukin-2 mutants
US8124066B2 (en) 2001-08-13 2012-02-28 University Of Southern California Methods of using interleukin-2 mutants with reduced toxicity
US7803361B2 (en) 2001-08-13 2010-09-28 University Of Southern California Therapeutic use of interleukin-2 mutants
EP2298354A2 (en) 2001-10-10 2011-03-23 BioGeneriX AG Remodelling and glycoconjugation of interferon-beta
EP2305313A2 (en) 2001-10-10 2011-04-06 BioGeneriX AG Remodelling and glycoconjugation of interferon-alpha (IFNa)
US7265085B2 (en) 2001-10-10 2007-09-04 Neose Technologies, Inc. Glycoconjugation methods and proteins/peptides produced by the methods
US7696163B2 (en) 2001-10-10 2010-04-13 Novo Nordisk A/S Erythropoietin: remodeling and glycoconjugation of erythropoietin
US7265084B2 (en) 2001-10-10 2007-09-04 Neose Technologies, Inc. Glycopegylation methods and proteins/peptides produced by the methods
US7226903B2 (en) 2001-10-10 2007-06-05 Neose Technologies, Inc. Interferon beta: remodeling and glycoconjugation of interferon beta
EP2080525A1 (en) 2001-10-10 2009-07-22 BioGeneriX AG Remodeling and Glycoconjugation of Peptides
EP2298354A3 (en) * 2001-10-10 2011-06-08 BioGeneriX AG Remodelling and glycoconjugation of interferon-beta
US7297511B2 (en) 2001-10-10 2007-11-20 Neose Technologies, Inc. Interferon alpha: remodeling and glycoconjugation of interferon alpha
US7214660B2 (en) 2001-10-10 2007-05-08 Neose Technologies, Inc. Erythropoietin: remodeling and glycoconjugation of erythropoietin
US7795210B2 (en) 2001-10-10 2010-09-14 Novo Nordisk A/S Protein remodeling methods and proteins/peptides produced by the methods
EP2042196A2 (en) 2001-10-10 2009-04-01 Neose Technologies, Inc. Remodelling and glycoconjugation of Granulocyte Colony Stimulating Factor (G-CSF)
US7399613B2 (en) 2001-10-10 2008-07-15 Neose Technologies, Inc. Sialic acid nucleotide sugars
US7179617B2 (en) 2001-10-10 2007-02-20 Neose Technologies, Inc. Factor IX: remolding and glycoconjugation of Factor IX
EP2279753A2 (en) 2001-10-10 2011-02-02 Novo Nordisk A/S Remodeling and glycoconjugation of peptides
EP2279755A2 (en) 2001-10-10 2011-02-02 BioGeneriX AG Remodelling and glycoconjugation of Fibroblast Growth Factor (FGF)
EP2279754A2 (en) 2001-10-10 2011-02-02 BioGeneriX AG Remodelling and glycoconjugation of human growth hormone (hGH)
US7416858B2 (en) 2001-10-10 2008-08-26 Neose Technologies, Inc. Pharmaceutical compositions of glycoconjugates
EP2305311A2 (en) 2001-10-10 2011-04-06 BioGeneriX AG Glycoconjugation of peptides
US7276475B2 (en) 2001-10-10 2007-10-02 Neose Technologies, Inc. Remodeling and glycoconjugation of peptides
EP2305312A2 (en) 2001-10-10 2011-04-06 BioGeneriX AG Remodelling and glycoconjugation of follicle-stimulating hormone (FSH)
EP2305314A2 (en) 2001-10-10 2011-04-06 BioGeneriX AG Remodelling and glycoconjugation of antibodies
US7173003B2 (en) 2001-10-10 2007-02-06 Neose Technologies, Inc. Granulocyte colony stimulating factor: remodeling and glycoconjugation of G-CSF
US7138371B2 (en) 2001-10-10 2006-11-21 Neose Technologies, Inc Remodeling and glycoconjugation of peptides
US7439043B2 (en) 2001-10-10 2008-10-21 Neose Technologies, Inc. Galactosyl nucleotide sugars
EP2322229A2 (en) 2001-10-10 2011-05-18 Novo Nordisk A/S Remodelling and glycoconjugation of Granulocyte Colony Stimulating Factor (G-CSF)
US7125843B2 (en) 2001-10-19 2006-10-24 Neose Technologies, Inc. Glycoconjugates including more than one peptide
US7473680B2 (en) 2001-11-28 2009-01-06 Neose Technologies, Inc. Remodeling and glycoconjugation of peptides
US7157277B2 (en) 2001-11-28 2007-01-02 Neose Technologies, Inc. Factor VIII remodeling and glycoconjugation of Factor VIII
US8017733B2 (en) 2002-01-18 2011-09-13 Biogen Idec Ma Inc. Polyalkylene polymer compounds and uses thereof
US8524660B2 (en) 2002-01-18 2013-09-03 Biogen Idec Ma Inc. Polyalkylene polymer compounds and uses thereof
US9125880B2 (en) 2002-12-26 2015-09-08 Mountain View Pharmaceuticals, Inc. Polymer conjugates of interferon-beta with enhanced biological potency
CN102319437B (zh) * 2002-12-26 2017-10-13 山景医药公司 具有增强的生物学效用的干扰素‑β的聚合物缀合物
CN102319437A (zh) * 2002-12-26 2012-01-18 山景医药公司 具有增强的生物学效用的干扰素-β的聚合物缀合物
US7803777B2 (en) 2003-03-14 2010-09-28 Biogenerix Ag Branched water-soluble polymers and their conjugates
US8247381B2 (en) 2003-03-14 2012-08-21 Biogenerix Ag Branched water-soluble polymers and their conjugates
US8063015B2 (en) 2003-04-09 2011-11-22 Novo Nordisk A/S Glycopegylation methods and proteins/peptides produced by the methods
US8853161B2 (en) 2003-04-09 2014-10-07 Novo Nordisk A/S Glycopegylation methods and proteins/peptides produced by the methods
EP2055189A1 (en) 2003-04-09 2009-05-06 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
US7691603B2 (en) 2003-04-09 2010-04-06 Novo Nordisk A/S Intracellular formation of peptide conjugates
EP2338333A2 (en) 2003-04-09 2011-06-29 BioGeneriX AG Glycopegylation methods and proteins/peptides produced by the methods
US9005625B2 (en) 2003-07-25 2015-04-14 Novo Nordisk A/S Antibody toxin conjugates
WO2005019260A1 (ja) * 2003-08-25 2005-03-03 Toray Industries, Inc. インターフェロンβ複合体
US7691975B2 (en) 2003-08-25 2010-04-06 Toray Industries, Inc. Interferon-β complex
US8916360B2 (en) 2003-11-24 2014-12-23 Novo Nordisk A/S Glycopegylated erythropoietin
US7405198B2 (en) 2003-11-24 2008-07-29 Neose Technologies, Inc. Glycopegylated erythropoietin
US9023992B2 (en) 2004-05-04 2015-05-05 Novo Nordisk Healthcare Ag Hydrophobic interaction chromatography purification of factor VII polypeptides
US10844110B2 (en) 2004-05-04 2020-11-24 Novo Nordisk Healthcare Ag O-linked glycoforms of polypeptides and method to manufacture them
US8791066B2 (en) 2004-07-13 2014-07-29 Novo Nordisk A/S Branched PEG remodeling and glycosylation of glucagon-like peptide-1 [GLP-1]
US8268967B2 (en) 2004-09-10 2012-09-18 Novo Nordisk A/S Glycopegylated interferon α
US10874714B2 (en) 2004-10-29 2020-12-29 89Bio Ltd. Method of treating fibroblast growth factor 21 (FGF-21) deficiency
US9200049B2 (en) 2004-10-29 2015-12-01 Novo Nordisk A/S Remodeling and glycopegylation of fibroblast growth factor (FGF)
US9029331B2 (en) 2005-01-10 2015-05-12 Novo Nordisk A/S Glycopegylated granulocyte colony stimulating factor
EP2514757A2 (en) 2005-01-10 2012-10-24 BioGeneriX AG Glycopegylated granulocyte colony stimulating factor
US9187546B2 (en) 2005-04-08 2015-11-17 Novo Nordisk A/S Compositions and methods for the preparation of protease resistant human growth hormone glycosylation mutants
EP2386571A2 (en) 2005-04-08 2011-11-16 BioGeneriX AG Compositions and methods for the preparation of protease resistant human growth hormone glycosylation mutants
US10160958B2 (en) 2005-04-11 2018-12-25 Horizon Pharma Rheumatology Llc Variant forms of urate oxidase and use thereof
US10731139B2 (en) 2005-04-11 2020-08-04 Horizon Pharma Rheumatology Llc Variant forms of urate oxidase and use thereof
US11345899B2 (en) 2005-04-11 2022-05-31 Horizon Therapeutics Usa, Inc. Variant forms of urate oxidase and use thereof
US11781119B2 (en) 2005-04-11 2023-10-10 Horizon Therapeutics Usa, Inc. Variant forms of urate oxidase and use thereof
EP2975135A1 (en) 2005-05-25 2016-01-20 Novo Nordisk A/S Glycopegylated factor IX
US8911967B2 (en) 2005-08-19 2014-12-16 Novo Nordisk A/S One pot desialylation and glycopegylation of therapeutic peptides
US8841439B2 (en) 2005-11-03 2014-09-23 Novo Nordisk A/S Nucleotide sugar purification using membranes
EP1939216A1 (en) * 2006-03-27 2008-07-02 Smirnov, Mikhail Nikolaevich Immunomodulating composition
EP1882744A1 (en) * 2006-03-27 2008-01-30 Smirnov, Mikhail Nikolaevich Method for producing an interleukin-2 preparation and the thus obtainable preparation
EP1882744A4 (en) * 2006-03-27 2009-03-25 Smirnov Mikhail Nikolaevich PROCESS FOR PRODUCTION OF INTERLEUKIN-2 PREPARATION AND PREPARATION OBTAINED BY SAID METHOD
EP1939216A4 (en) * 2006-03-27 2009-04-01 Smirnov Mikhail Nikolaevich IMMUNOMODULATORY COMPOSITION
US9187532B2 (en) 2006-07-21 2015-11-17 Novo Nordisk A/S Glycosylation of peptides via O-linked glycosylation sequences
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
US9050304B2 (en) 2007-04-03 2015-06-09 Ratiopharm Gmbh Methods of treatment using glycopegylated G-CSF
US9493499B2 (en) 2007-06-12 2016-11-15 Novo Nordisk A/S Process for the production of purified cytidinemonophosphate-sialic acid-polyalkylene oxide (CMP-SA-PEG) as modified nucleotide sugars via anion exchange chromatography
DE202008017456U1 (de) 2007-08-27 2009-08-27 Biogenerix Ag Flüssig-Formulierung von G-CSF-Konjugaten
EP2578235A2 (en) 2007-08-27 2013-04-10 BioGeneriX AG Liquid formulation of G-CSF conjugate
EP2933264A2 (en) 2008-01-22 2015-10-21 Araim Pharmaceuticals, Inc. Tissue protective peptides and peptide analogs for preventing and treating diseases and disorders associated with tissue damage
US9150848B2 (en) 2008-02-27 2015-10-06 Novo Nordisk A/S Conjugated factor VIII molecules
EP2672259A1 (en) 2008-05-13 2013-12-11 Advanced Liquid Logic, Inc. Droplet actuator devices, systems and methods
EP2672260A1 (en) 2008-05-13 2013-12-11 Advanced Liquid Logic, Inc. Droplet actuator devices, systems and methods
US11982670B2 (en) 2009-06-25 2024-05-14 Horizon Therapeutics Usa, Inc. Methods and kits for predicting infusion reaction risk and antibody-mediated loss of response by monitoring serum uric acid during pegylated uricase therapy
US10139399B2 (en) 2009-06-25 2018-11-27 Horizon Pharma Rheumatology Llc Methods and kits for predicting infusion reaction risk and antibody-mediated loss of response by monitoring serum uric acid during PEGylated uricase therapy
US11639927B2 (en) 2009-06-25 2023-05-02 Horizon Therapeutics Usa, Inc. Methods and kits for predicting infusion reaction risk and antibody-mediated loss of response by monitoring serum uric acid during PEGylated uricase therapy
US10823727B2 (en) 2009-06-25 2020-11-03 Horizon Pharma Rheumatology Llc Methods and kits for predicting infusion reaction risk and antibody-mediated loss of response by monitoring serum uric acid during pegylated uricase therapy
US11598767B2 (en) 2009-06-25 2023-03-07 Horizon Therapeutics Usa, Inc. Methods and kits for predicting infusion reaction risk and antibody-mediated loss of response by monitoring serum uric acid during pegylated uricase therapy
US10960079B2 (en) 2010-11-12 2021-03-30 Nektar Therapeutics Conjugates of an IL-2 moiety and a polymer
US20210023230A1 (en) * 2010-11-12 2021-01-28 Nektar Therapeutics Conjugates of an il-2 moiety and a polymer
US11091525B2 (en) 2010-11-12 2021-08-17 Nektar Therapeutics Method of refolding an interleukin-2 (IL-2) protein
WO2012107417A1 (en) 2011-02-10 2012-08-16 Roche Glycart Ag Mutant interleukin-2 polypeptides
EP3489255A1 (en) 2011-02-10 2019-05-29 Roche Glycart AG Mutant interleukin-2 polypeptides
EP3971206A1 (en) 2011-02-10 2022-03-23 Roche Glycart AG Mutant interleukin-2 polypeptides
WO2013129549A1 (ja) 2012-02-29 2013-09-06 東レ株式会社 体腔液貯留抑制剤
WO2013156488A2 (en) 2012-04-16 2013-10-24 Leverton Licence Holdings Limited Optimised subcutaneous therapeutic agents
US11351112B2 (en) 2012-04-16 2022-06-07 Cantab Biopharmaceuticals Patents Limited Optimised subcutaneous therapeutic agents
US9844582B2 (en) 2012-05-22 2017-12-19 Massachusetts Institute Of Technology Synergistic tumor treatment with extended-PK IL-2 and therapeutic agents
WO2013177187A3 (en) * 2012-05-22 2014-01-16 Massachusetts Institute Of Technology Synergistic tumor treatment with extended-pk il-2 and therapeutic agents
US20150017120A1 (en) * 2013-06-13 2015-01-15 Massachusetts Institute Of Technology Synergistic tumor treatment with extended-pk il-2 and adoptive cell therapy
US10864253B2 (en) 2016-04-29 2020-12-15 Araim Pharmaceuticals, Inc. Tissue protective peptides for preventing and treating diseases and disorders associated with tissue damage
WO2018184965A1 (en) 2017-04-03 2018-10-11 F. Hoffmann-La Roche Ag Immunoconjugates of il-2 with an anti-pd-1 and tim-3 bispecific antibody
WO2018184964A1 (en) 2017-04-03 2018-10-11 F. Hoffmann-La Roche Ag Immunoconjugates of an anti-pd-1 antibody with a mutant il-2 or with il-15
AU2018394189B2 (en) * 2017-12-27 2023-12-21 Kyowa Kirin Co., Ltd. IL-2 variant
WO2020158691A1 (ja) 2019-01-28 2020-08-06 東レ株式会社 肝細胞増殖因子又はその活性断片のポリエチレングリコール修飾体
WO2020158690A1 (ja) 2019-01-28 2020-08-06 東レ株式会社 肝細胞増殖因子又はその活性断片のポリエチレングリコール修飾体
WO2021001289A1 (en) 2019-07-02 2021-01-07 F. Hoffmann-La Roche Ag Immunoconjugates comprising a mutant interleukin-2 and an anti-cd8 antibody
WO2021209402A2 (en) 2020-04-15 2021-10-21 F. Hoffmann-La Roche Ag Immunoconjugates
WO2021231278A1 (en) 2020-05-11 2021-11-18 F. Hoffmann-La Roche Ag Combination therapy with modified pbmcs and an immunoconjugate
WO2022117692A2 (en) 2020-12-04 2022-06-09 F. Hoffmann-La Roche Ag Ph-dependent mutant interleukin-2 polypeptides
WO2022148853A1 (en) 2021-01-11 2022-07-14 F. Hoffmann-La Roche Ag Immunoconjugates
WO2023052541A1 (en) 2021-09-30 2023-04-06 Imcheck Therapeutics Combination of an anti-btn3a activating antibody and an il-2 agonist for use in therapy
WO2023062050A1 (en) 2021-10-14 2023-04-20 F. Hoffmann-La Roche Ag New interleukin-7 immunoconjugates
WO2023062048A1 (en) 2021-10-14 2023-04-20 F. Hoffmann-La Roche Ag Alternative pd1-il7v immunoconjugates for the treatment of cancer

Also Published As

Publication number Publication date
DK97987A (da) 1987-02-25
EP0229108A1 (en) 1987-07-22
IN163200B (US20100223739A1-20100909-C00025.png) 1988-08-20
FI93424C (fi) 1995-04-10
ZA864766B (en) 1988-02-24
IE861706L (en) 1986-12-26
IL79235A (en) 1991-01-31
DE3676670D1 (de) 1991-02-07
PT82834B (pt) 1988-12-15
DK169874B1 (da) 1995-03-20
IE59406B1 (en) 1994-02-23
PT82834A (en) 1986-07-01
GR861641B (en) 1986-09-12
KR870000423A (ko) 1987-02-18
MX174442B (es) 1994-05-17
JPS62503171A (ja) 1987-12-17
FI93424B (fi) 1994-12-30
DK97987D0 (da) 1987-02-25
EP0229108B1 (en) 1990-12-27
NZ216618A (en) 1989-05-29
FI870809A (fi) 1987-02-25
CA1291708C (en) 1991-11-05
JP2524586B2 (ja) 1996-08-14
IL79235A0 (en) 1986-09-30
FI870809A0 (fi) 1987-02-25
PH25004A (en) 1991-01-28
KR900004801B1 (ko) 1990-07-06

Similar Documents

Publication Publication Date Title
US4917888A (en) Solubilization of immunotoxins for pharmaceutical compositions using polymer conjugation
EP0229108B1 (en) Solubilization of proteins for pharmaceutical compositions using polymer conjugation
US4766106A (en) Solubilization of proteins for pharmaceutical compositions using polymer conjugation
EP0402378B1 (en) Conjugation of polymer to colony stimulating factor-1
US5153265A (en) Conjugation of polymer to colony stimulating factor-1
AU626518B2 (en) Solubilization of proteins for pharmaceutical compositions using polyproline conjugation
US4745180A (en) Solubilization of proteins for pharmaceutical compositions using heparin fragments
EP0247860B1 (en) Tumor necrosis factor formulation and its preparation
Pasut et al. Protein, peptide and non-peptide drug PEGylation for therapeutic application
US5711944A (en) Interferon polymer conjugates
CA2617064A1 (en) Conjugates of a g-csf moiety and a polymer
JP2010184929A (ja) ポリオール−ifn−ベータ複体
AU580431B2 (en) Solubilization of proteins for pharmaceutical compositions using polymer conjugation
PT89861B (pt) Processo para a conjugacao de polimeros com factor estimulador de colonias-1
KR20040086521A (ko) 생물학적 활성물질과 생체적합성 고분자의 1:1 접합체,이의 제조방법과 이를 함유하는 약학 조성물

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AU DK FI JP NO

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): AT BE CH DE FR GB IT LU NL SE

WWE Wipo information: entry into national phase

Ref document number: 870809

Country of ref document: FI

WWE Wipo information: entry into national phase

Ref document number: 1986903990

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 1986903990

Country of ref document: EP

WWG Wipo information: grant in national office

Ref document number: 1986903990

Country of ref document: EP

WWG Wipo information: grant in national office

Ref document number: 870809

Country of ref document: FI