WO1997003106A1 - Poly(ethylene glycol) and related polymers monosubstituted with propionic or butanoic acids and functional derivatives thereof for biotechnical applications - Google Patents

Poly(ethylene glycol) and related polymers monosubstituted with propionic or butanoic acids and functional derivatives thereof for biotechnical applications Download PDF

Info

Publication number
WO1997003106A1
WO1997003106A1 PCT/US1996/011261 US9611261W WO9703106A1 WO 1997003106 A1 WO1997003106 A1 WO 1997003106A1 US 9611261 W US9611261 W US 9611261W WO 9703106 A1 WO9703106 A1 WO 9703106A1
Authority
WO
WIPO (PCT)
Prior art keywords
peg
poly
ppm
group
thiol
Prior art date
Application number
PCT/US1996/011261
Other languages
French (fr)
Inventor
J. Milton Harris
Antoni Kozlowski
Original Assignee
Shearwater Polymers, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Shearwater Polymers, Inc. filed Critical Shearwater Polymers, Inc.
Priority to AU63457/96A priority Critical patent/AU6345796A/en
Publication of WO1997003106A1 publication Critical patent/WO1997003106A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C08ORGANIC MACROMOLECULAR COMPOUNDS; THEIR PREPARATION OR CHEMICAL WORKING-UP; COMPOSITIONS BASED THEREON
    • C08GMACROMOLECULAR COMPOUNDS OBTAINED OTHERWISE THAN BY REACTIONS ONLY INVOLVING UNSATURATED CARBON-TO-CARBON BONDS
    • C08G65/00Macromolecular compounds obtained by reactions forming an ether link in the main chain of the macromolecule
    • C08G65/02Macromolecular compounds obtained by reactions forming an ether link in the main chain of the macromolecule from cyclic ethers by opening of the heterocyclic ring
    • C08G65/32Polymers modified by chemical after-treatment
    • C08G65/329Polymers modified by chemical after-treatment with organic compounds

Definitions

  • the invention relates to derivatives of poly(ethylene glycol) and related hydrophilic polymers, to methods for their synthesis, and to surfaces and molecules modified by these polymers for biotechnical use.
  • PEG poly(ethylene glycol)
  • PEO poly(ethylene oxide)
  • alpha-, omega-dihydroxy1 poly(ethylene glycol) can be represented in brief form as HO-PEG-OH where it is understood that the -PEG- sy bol represents the following structural unit:
  • PEG is soluble in organic solvents.
  • PEG attached to enzymes can result in PEG-enzyme conjugates that are soluble and active in organic solvents. Attachment of PEG to protein can reduce the immunogenicity and rate of kidney clearance of the PEG-protein conjugate as compared to the unmodified protein, which may result in dramatically increased blood circulation lifetimes for the conjugate.
  • PEG attached to surfaces can reduce protein and cell adsorption to the surface and alter the electrical properties of the surface.
  • PEG attached to liposomes can result in a great increase in the blood circulation lifetime of these particles and thereby possibly increase their utility for drug delivery.
  • Harris J.M. Harris, Ed., "Biomedical and Biotechnical Applications of Polyethylene Glycol Chemistry, " Plenum, New York, 1992) .
  • Chemically active or "activated" derivatives of the PEG polymer are prepared to attach the PEG to molecules and surfaces.
  • a number of such derivatives have been prepared, some of which are of somewhat general utility, some of which are directed to solving specific problems associated with particular applications, and some of which have not proved useful or have problems that limit their usefulness.
  • active esters of alpha-, omega-dicarboxylic acids of PEG have been prepared, typically for attachment of PEG to amino groups .
  • an active ester is poly(ethylene glycol) di - succinimidyl succinate (symbolized in shorthand as "SS- PEG-SS”) : H0 2 C-CH 2 CH 2 - C0 2 -PEG-0 2 C- CH 2 CH 2 - C0 2 -H + 2NHS -OH ⁇ ⁇ NHS-0 2 C-CH 2 CH 2 - C0 2 - PEG-0 2 C- CH 2 CH 2 - C0 2 -NHS
  • N-hydroxy succinimide from which the succinimidyl active ester moiety is derived.
  • N-hydroxylsuccinimide active ester moiety will be represented as -C0 2 -NHS.
  • the SS-PEG-SS active ester reacts rapidly with amino groups on proteins and other molecules to form a stable amide linkage (-C0-NH-) .
  • a problem with the active ester is that SS-PEG-SS possesses ester linkages in the backbone that remain intact after coupling to an amine such as a protein (represented as PRO-NH 2 ) :
  • the intact ester linkage that remains after the protein and PEG conjugate is formed is subject to hydrolysis.
  • the PEG detaches from the modified protein in aqueous solution.
  • the rate at which hydrolysis occurs can vary with the environment .
  • U.S. Patent No. 4,670,417 discloses hemoglobin modified with various dicarboxylic acid PEGs in which the ester linkage that would otherwise remain in the conjugate is replaced with an ether linkage.
  • the hemoglobin is modified in the presence of amino acids or amines that are said to also react with the dicarboxylic acid PEG to prevent excessive carboxyl groups from reacting with the hemoglobin, to avoid crosslinking and gelation of the reaction mixture, and to avoid the time, expense, and use of large reaction vessels that would otherwise be required.
  • special precautions are necessary to prevent cross linking with this difunctional material.
  • PEGs that are activated at each terminus with the same reactive moiety have been referred to in the art as being "homobifunctional" to indicate that the active moieties at the polymer ends are normally the same.
  • PEG derivatives in which different functional groups are present at the two terminae of the polymer chain. These derivatives are sometimes referred to as heterobifunctional or heterofunctional .
  • PEG derivatives active on one end and capped on the opposite end by a relatively nonreactive methoxy group have been prepared. These mPEGs are typically considered to be monofunctional, so that only one end of the PEG is active for conjugation to other substances. For example, one PEG is frequently used in a form in which there is a relatively nonreactive methoxy group at one terminus and the reactive succinimidyl succinate group at the other terminus :
  • the invention is based upon the recognition that many of the activated carboxylic acid PEGs have poor reactivity and are either too slow or too fast to be generally useful for conjugation.
  • the NHS ester of carboxymethylated PEG is so reactive that it hydrolyzes almost immediately upon solution in water. This high reactivity is a serious deficiency of PEG derivatives based on carboxymethylated PEG.
  • the invention provides heterofunctional and monofunctional PEGs and related polymers having a single carboxylic acid moiety of suitable reactivity, which can be activated.
  • the active esters of these polymer acids have a single active ester and no other ester linkages.
  • the active esters have a half life in water of from about 10 to 25 minutes.
  • These esters include either a propionic or butanoic acid moiety and a polymer such as PEG or a related polymer.
  • These related polymers include poly(alkylene oxides) , poly(oxyethylated polyols), poly(olefinic alcohols) , and poly(acrylomorpholine) .
  • conjugates of these activated polymers with biologically active substances such as polypeptides, proteins, enzymes, phospholipids, lipids, liposomes, nucleosides, oligonucleotides, drugs, dyes, and the surfaces of solid materials that are compatible with living organisms, tissue, or fluids, which are sometimes referred to as biomaterials. Further provided are methods for preparation of these activated polymers and conjugates.
  • one terminus of PEG is the activated propionic acid or butanoic acid moiety and the other terminus can be capped with a relatively nonreactive methoxy group, for example, or can be activated with any other functional group, which may be protected or unprotected.
  • the succinimidyl ester of heterofunctional PEG propionic acid can be represented as follows :
  • X- can include any activating group other than a carboxylic acid or activated carboxylic acid or can be a relatively nonreactive group such as methoxy, CH 3 0- .
  • "X-" can include the thiol-selective vinyl sulfone and other sulfone moieties as shown in commonly assigned U.S. Patent No. 5,446,090, which issued on August 29, 1995, the contents of which are incorporated herein by reference.
  • the utility of the heterofunctional and monofunctional propionic and butanoic acid PEGs is believed to come from four sources.
  • succinimidyl ester of PEG propionic acid has ideal reactivity for attachment to amines, such as proteins, in aqueous solution.
  • the succinimidyl ester of PEG butanoic acid is less reactive than the propionic acid, but is still of utility.
  • the reactions at the terminae of the heterofunctional and monofunctional PEG molecules can be controlled to limit cross linking generally more easily than typically is true of homobifunctional derivatives.
  • the hydrolytic stability of the ether linkages in the backbone of PEG propionic acid results in stable conjugates after chemical coupling to another molecule or surface.
  • the hydrolytic stability of the ether linkage in the intermediate heterofunctional and monofunctional propionic acid PEGs permits ready purification of the intermediate by ion exchange chromatography in aqueous medium.
  • the structure of the active esters of poly(ethylene glycol) of the invention can be represented as follows :
  • the moiety (OCH 2 CH 2 ) n represents PEG wherein n is from about 20 to 4000. More typically, n is from 20 to
  • Z is selected from hydrolytically stable groups including -O- , -S-, -NHCO-, -CONH-,
  • m is from 2 to 3 for propionic and butanoic acid derivatives, respectively.
  • Q is selected from the group including hydrogen, tert- butyl, N-succinimide, N-sulfosuccinimide, N-phthalimide, N-glutarimide, N-tetrahydrophthalimide, N-norbornene-2, 3-dicarboximide, hydroxybenzotriazole, and hydroxy-7-azabenzotriazole.
  • the invention provides active esters that have suitable reactivity for modification of other molecules and surfaces. Use of these
  • Example 1 Synthesis of CH 3 0- (CH 2 CH 2 0) n -CH 2 CH 2 C0 2 -NHS
  • Example 2 Synthesis of CH 3 0- (CH 2 CH 2 0) n -CH 2 CH 2 -S- CH 2 CH 2 C0 2 -NHS
  • Example 3 Synthesis of CH 3 0- (CH,CH 2 0) n -CH 2 CH 2 CH 2 C0 2 -NHS
  • Example 4 - Rates of hydrolysis of PEG-NHS active esters
  • Example 5 Synthesis of HO- (CH-,CH 2 0) n -CH 2 CH 2 -S-CH 2 CH 2 -C0 2 H
  • Example 6 Synthesis of HO- (CH 2 CH 2 0) n -CH 2 CH 2 -S-CH 2 CH 2
  • Example 14 Synthesis of (CH 3 ) -C-O-CONH- (CH 2 CH 2 0) n -CH 2 CH 2 C00H
  • Example 15 - ⁇ -methoxy- ⁇ -dipalmitoylphos- phatidylethanolamide of propionic acid of PEG (a"PEG-phospholipid)
  • Example 16 - ⁇ -methoxy- ⁇ -distearoyl- phosphatidylethanolamide of propionic acid of PEG (a "PEG"-phospholipid)
  • Example 17 Synthesis of HS-CH 2 CH 2 CONH- (CH 2 CH 2 0) n -CH 2 CH 2 -S-CH 2 CH 2 C0 2 H
  • Example 18 Coupling of methoxy-PEG-SPA to glass surfaces
  • Example 19 Coupling of PEG-SPA to proteins
  • M-PEG-5000-methanesulfonate from Shearwater Polymers, 70.0 g; 0.0206 moles was added to a mixture of 280 ml toluene and 420 ml absolute ethanol and stirred to dissolve.
  • Ethyl-3-mercaptopropionoate (3x excess; 7.84 ml; 8.15 g; 0.061 moles) was added to -li ⁇ the reaction via syringe and the reaction was heated under a nitrogen atmosphere to 60°C for three hours. The reaction mixture was cooled to room temperature, filtered to remove insoluble salts and concentrated to about 250 ml under reduced pressure. This is added to 1200 ml cold diethyl ether, giving a pale yellow precipitate which is dried in vacuo overnight .
  • M-PEG-5000-S-CH 2 CH 2 -COOC 2 H S from the preceding step (70.0 g; 0.0206 moles) was added to 840 ml distilled deionized water and stirred to dissolve.
  • sodium hydroxide (1.4 g) was added to 35 ml distilled deionized water and stirred to dissolve.
  • the sodium hydroxide solution was added to the PEG solution until the pH was 12-13.
  • the solution was then stirred at room temperature for one hour.
  • Oxalic acid was added to adjust the pH to 3.
  • the solution was extracted with CH 2 C1 2 three times (200/200/200 ml) .
  • M-PEG-5000-COOH from the preceding step (3.00 g; 0.8823 mmol)
  • N,N' dicyclohexyl carbodiimide 1.5x excess; 0.2731 g; 0.001324 mmol
  • N-hydroxysuccinimide 1.5x excess; 0.1523 g; 0.001324 mmol
  • M-PEG methanesulfonate (20 g, 1 equivalent, MW 5000 daltons, from Shearwater Polymers) was dissolved in 50 ml of toluene and added to the above mixture. The resulting mixture was reflexed overnight. The reaction mixture was then concentrated to half its original volume, extracted with 15 ml of 10% aqueous NaCl solution, extracted with 10 ml of 1% aqueous hydrochloric acid, and the aqueous extracts combined. The collected aqueous layers were extracted with dichloromethane (50 ml x 3) , and the organic layer was dried with magnesium sulfate for 3 hours, filtered, and evaporated to dryness. Yield: 20 g of PEG malonic ester.
  • M-PEG malonic ester (18 g) was dissolved in 240 ml of IN sodium hydroxide containing 12 g of sodium chloride, and the mixture was stirred for one hour.
  • M-PEG malonic acid (15 g) was dissolved in 120 ml of dioxane and refluxed for 8 hours, then concentrated to dryness. The residue was dissolved in 100 ml water, extracted with dichloromethane (70 ml and 50 ml) , dried over magnesium sulfate, and the solution concentrated by rotary evaporation. The residue was precipitated by addition to cold diethyl ether. Yield: 11 g.
  • SCM-PEG (10) has a half life of 0.75 minutes and reacts with water too quickly to be useful for conjugation with biologically active substances or surfaces.
  • SS-PEG (6) with a half life of 9.8 minutes, has good reactivity, but contains an ester linkage and forms hydrolytically unstable conjugates.
  • SSPA-PEG (5) which is prepared in accordance with the present invention, has good reactivity and no ester linkage that could interfere with hydrolytic stability of conjugates.
  • SPA-PEG (4) has ideal reactivity at a half life of 16.5 minutes and no ester linkage that could interfere with hydrolytic stability. SBA-PEG (1) reacts more slowly, at a half life of 23.3 minutes, but still is of utility. Generally, a half life of 10 to about 20 or 25 minutes is desirable.
  • PEG-3400-methanesulfonate (70.0 g; 11% substituted; 0.0206 moles, Shearwater Polymers) was added to a mixture of 280 ml toluene and 420 ml absolute ethanol and stirred to dissolve.
  • Sodium hydroxide (3x excess; 2.32 g; 0.0581 moles) was dissolved in 56 ml absolute ethanol and added to the PEG-mesylate.
  • Ethyl-3- mercaptopropionoate (3x excess; 7.84 ml; 8.15 g; 0.0607 moles) was added to the reaction via syringe and the reaction was heated under a nitrogen atmosphere to 60°C for three hours.
  • the reaction mixture is cooled to room temperature, filtered to remove insoluble salts and concentrated to about 250 ml under reduced pressure. This is added to 1200 ml cold diethyl ether, giving the desired ester as a pale yellow precipitate which is dried in vacuo overnight .
  • the extract was dried over Na 2 S0 4 /MgS0 4 , filtered, concentrated under reduced pressure to about 150 ml and added to 1000 ml cold diethyl ether to precipitate the product .
  • the product was dried in vacuo overnight . Yield: 61.6 g, 88%.
  • the mixture was purified by ion exchange chromatography on Sepharose FF (Pharmacia) .
  • -NMR DMSO-d 2.63 ppm (m, -SCH 2 CH 2 -, 4H) , 3.50 ppm (s, PEG backbone, 304H) , 4.57 ppm (t, OH, IH) .
  • CH 2 CH-C0 2 - (CH 2 CH 2 0) n CH 2 CH 2 S-CH 2 CH 2 -C0 2 -NHS
  • this product (0.946 g; 0.278 mmol), N,N' dicyclohexyl carbodiimide (1.5x excess; 0.0861 g; 0.417 mmol) and N-hydroxysuccinimide (1.5x excess; 0.0480 g; 0.417 mmol) were dissolved in 30.0 ml CH 2 C1 2 .
  • the flask was immersed in an ice bath and stirred overnight. The next day the reaction mixture was filtered, concentrated under reduced pressure, filtered and precipitated into cold diethyl ether. Yield 0.592 g., 63%.
  • the reaction was heated to reflux and the methanol was collected in the Dean- Stark trap along with an additional 8 ml solvent.
  • the reaction mixture was cooled under nitrogen to room temperature, during which time the color changed from pale yellow to deep red-brown. Allyl chloride (1.35 g; 1.43 ml; 0.00176 moles) was added via syringe and the mixture was stirred overnight at room temperature. The following morning the mixture was filtered through a buchner funnel, concentrated under reduced pressure and precipitated into cold ether. Yield 49.17 g, 82%.
  • the isolated product was checked by NMR to confirm that substitution had occurred.
  • allyl-PEG 3400 (49.17 g; 0.0144 moles) was dissolved in 250 ml toluene and azeotropically dried approximately 15 minutes to remove 15 ml cloudy solvent/water.
  • Polymers 100 . 0 g ; 0 . 0281 moles ) was added to a mixture of 400 ml toluene and 300 ml absolute ethanol and stirred to dissolve .
  • Ethyl - 3 -mercaptopropionate ( 6 . 0 g; 0.0445 moles. 79.2% of stoichiometric amount) and sodium hydroxide (1.6 g; 0.04 moles, 71.1% of stoichiometric amount) dissolved in 40 ml absolute ethanol were added to the reaction mixture.
  • the reaction was heated under nitrogen atmosphere at 60°C for three hours.
  • the reaction mixture was cooled to room temperature, filtered to remove insoluble salt and concentrated to about 300 ml under reduced pressure.
  • the reaction product (mixture of a , ⁇ - diamino of PEG, ⁇ -amino- ⁇ -thiopropionic acid of PEG, and ⁇ , ⁇ -dithiopropionic acid of PEG) was extracted with dichloromethane (3 X 300 ml) . The extract was dried over anhydrous magnesium sulfate, filtered and concentrated to dryness. Pure ⁇ -amino-hydrochloride- ⁇ - thiopropionic acid of PEG was separated by ion exchange chromatography on S-Sepharose FF (Pharmacia) . Yield 37.5 g.
  • EXAMPLE 12 alpha-amino, omega-propionic acid of PEG H 2 NCH 2 CH 2 0 ( CH 2 CH 2 0 ) n CH 2 CH 2 C0 2 H
  • the pH of the solution was maintained with periodical addition of 0.1 N sodium hydroxide.
  • the solution was added to the mixture of 160 ml concentrated ammonium hydroxide and 16 g ammonium chloride and stirred 46 hours at room temperature.
  • the reaction product was extracted with dichloromethane (3 X 50 ml) .
  • the extract was washed with 20 ml 1 M hydrochloric acid, 20 ml distilled water and dried with anhydrous sodium sulfate.
  • the solvent was distilled under reduced pressure giving 4.2 g of ex ⁇ amine hydrochloride- ⁇ -propionic acid of PEG.
  • ⁇ -Methoxy- ⁇ -propionic acid succinimidyl ester of PEG of molecular weight 2170 (MSPA) (10 g, 0.00461 moles) was dissolved in chloroform (40 ml) and treated with solid distearoylphosphatidylethanolamine (DSPE) (3.72 g, 0.00497 moles) and triethylamine (2.4 ml) .
  • MSPA molecular weight 2170
  • DSPE solid distearoylphosphatidylethanolamine
  • triethylamine 2.4 ml
  • PEG amino acid ( ⁇ -aminohydrochloride- ⁇ -S- propionic acid, from Example 10) (1.83 grams) of molecular weight 400 was dissolved in 250 ml of benzene containing 1.0 ml of triethylamine and 20 ml of dry methylene chloride. Benzene was distilled off under reduced pressure. The residue was redissolved in 10 ml of acetonitrile, and succinimidyl-3- (2- pyridyldithio)propionate (SPDP) dissolved in 10 ml of acetonitrile was added. The mixture was stirred at room temperature under nitrogen atmosphere overnight . Gel permeation chromatography showed no aminoacid peak and a new monoacid peak. The solvent was distilled off under reduced pressure. Yield 3.2 g.
  • the product from the preceding step (3.2 g) was dissolved in 300 ml of distilled water containing 2.5 g of dithiothreitol and stirred at room temperature under nitrogen atmosphere for 3 hours .
  • the reaction mixture was applied to a DEAE Sepharose FF column (100 ml) , and the column was washed with 800 ml of distilled water.
  • the product was eluted with 200 ml of 0.5 M NaCl .
  • the pH of the eluate was adjusted to 3.0 with
  • Quartz slides were cleaned and activated for surface modification by soaking in 1% (w/w) aqueous NaOH at 90°C for 10 minutes, 3% (w/w) HCl at 90°C for 10 minutes, and boiling in 30% (w/w) H 2 0 : for 1 hour to remove trace organics, then rinsed with water.
  • the clean glass slides were prepared for functionalization with aminopropylsilane by drying under vacuum of IO "3 torr for 1 hour to remove excess surface water.
  • the glass slides were then exposed to a 2% (v/v) solution of silane in anhydrous toluene for 4 hours at room temperature.
  • the capillaries were rinsed with toluene and cured in a vacuum oven at 190°C, IO" 3 torr for 12 hours. This procedure gave quartz slides with available amino groups on the surface.
  • Methoxy-PEG-SPA from Example 1 was grafted to the functionally activated quartz surfaces by reacting as 5% (w/v) solution in 0.05 M sodium bicarbonate (pH 8.3) for 4 hours at 40°C. After PEG immobilization, surfaces were rinsed with toluene, dried under vacuum and rinsed with water. Examination with X-ray photoelectron spectroscopy (XPS) showed the presence of a large C-0 peak consistent with attachment of PEG. Also the water-contact angle is near zero, as expected for a PEG-coated surface. Finally, adsorption studies with fibrinogen revealed that fibrinogen adsorption on the PEG-coated surface has been reduced by approximately 98% relative to uncoated quartz.
  • XPS X-ray photoelectron spectroscopy
  • PEG-SPA Succinimidyl esters of PEG propionic acids
  • the enzyme subtilisin (2 ml of a 2.37 mg/ml solution) was coupled to acryloyi-PEG-SPA (from Example 7) (MW 3400, 10 mg) by reaction in 2 ml of borate buffer (0.1 M, pH 8.0) for one hour at 4°C.
  • the protein was purified by ultrafiltration with an Amicon PM 30 ultrafiltration membrane. Analysis with fluorescamine assay showed that two lysine groups had been modified by PEG attachment.
  • bovine alkaline phosphatase MW 140,000 was coupled to methoxy-PEG- SPA, MW 5000, (from Example 1) by reaction of 20 mg of enzy e and 30 mg of the PEG in 2 ml of buffer containing 0.2 M sodium phosphate and 0.5 M NaCl (pH 7.5) at 4°C for 30 minutes. Unreacted PEG was removed by ultrafiltration, as above. Fluorescamine analysis showed that 20% of the available lysines were modified. Analysis on size exclusion chromatography (Toya Soda TSK 3000 column with pH 7 phosphate buffer eluent) showed that the molecular weight of the PEG-protein conjugate was approximately 30 thousand daltons greater than that of native protein.
  • These other polymers include poiy(vinyl alcohol) (“PVA”) ; other poly(alkylene oxides) such as poly(propylene glycol) (“PPG”) and the like; and poly(oxyethylated polyols) such as poly(oxyethylated glycerol) , poly(oxyethylated sorbitol) , and poly(oxyethylated glucose) , and the like.
  • PVA poiy(vinyl alcohol)
  • PPG poly(propylene glycol)
  • poly(oxyethylated polyols) such as poly(oxyethylated glycerol) , poly(oxyethylated sorbitol) , and poly(oxyethylated glucose) , and the like.
  • the polymers can be homopolymers or random or block copolymers and terpolymers based on the monomers of the above polymers, straight chain or branched, or substituted or unsubstituted similar to mPEG and other capped, monofunctional PEGs having a single active site available for attachment to a linker.
  • suitable additional polymers include poly(oxazoline) , poly(acryloylmorpholine) (“PAcM”) as described in published Italian Patent Application MI-92-A-0002616 filed November 17, 1992, and poly(vinylpyrrolidone) (“PVP”) .
  • PVP and poly(oxazoline) are well known polymers in the art and their preparation and use in the syntheses described above for mPEG should be readily apparent to the skilled artisan.
  • the invention has been described with respect to several particular examples and embodiments. However, the foregoing examples and description are not intended to limit the invention to the exemplified embodiments, and the skilled artisan should recognize that variations can be made within the scope and spirit of the invention as described in the foregoing specification. The invention includes all alternatives, modifications, and equivalents that may be included within the true scope and spirit of the invention as defined by the appended claims.

Abstract

Active esters of PEG acids and related polymers are provided that have a single propionic or butanoic acid moiety and no other ester linkages. These polymer acids have a half life in water of from about 10 to 25 minutes. For example, alpha-methoxy, omega-propionic acid succinimidyl ester of PEG ('methoxy-PEG-SPA') has a nearly ideal reactivity with amino groups on proteins and other biologically active substances. The half life of methoxy-PEG-SPA is about 16.5 minutes in water. The invention also provides conjugates with proteins, enzymes, polypeptides, drugs, dyes, nucleosides, oligonucleotides, lipids, phospholipids, liposomes, and surfaces of solid materials that are compatible with living organisms, tissue, or fluid.

Description

POLY(ETHYLENE GLYCOL) AND RELATED POLYMERS MONOSUBSTITUTED WITH PROPIONIC OR BUTANOIC ACIDS AND FUNCTIONAL DERIVATIVES THEREOF FOR BIOTECHNICAL APPLICATIONS
Field of the Invention The invention relates to derivatives of poly(ethylene glycol) and related hydrophilic polymers, to methods for their synthesis, and to surfaces and molecules modified by these polymers for biotechnical use.
Background of the Invention Chemical attachment of the hydrophilic polymer poly(ethylene glycol) ("PEG") , which is also known as poly(ethylene oxide) ("PEO") , to molecules and surfaces is of great utility in biotechnology. In its most common form, PEG is a linear polymer terminated at each end with hydroxyl groups :
HO-CH2CH20- (CH2CH20)n-CH2CH2-OH
The above polymer, alpha-, omega-dihydroxy1 poly(ethylene glycol) can be represented in brief form as HO-PEG-OH where it is understood that the -PEG- sy bol represents the following structural unit:
-CH2CH20- (CH2CH20)n-CH2CH2- As an example of biotechnical applications of PEG, some active derivatives of PEG have been attached to proteins and enzymes with beneficial results. PEG is soluble in organic solvents. PEG attached to enzymes can result in PEG-enzyme conjugates that are soluble and active in organic solvents. Attachment of PEG to protein can reduce the immunogenicity and rate of kidney clearance of the PEG-protein conjugate as compared to the unmodified protein, which may result in dramatically increased blood circulation lifetimes for the conjugate.
PEG attached to surfaces can reduce protein and cell adsorption to the surface and alter the electrical properties of the surface. Similarly, PEG attached to liposomes can result in a great increase in the blood circulation lifetime of these particles and thereby possibly increase their utility for drug delivery. The above applications and many leading references are described in the book by Harris (J.M. Harris, Ed., "Biomedical and Biotechnical Applications of Polyethylene Glycol Chemistry, " Plenum, New York, 1992) .
Chemically active or "activated" derivatives of the PEG polymer are prepared to attach the PEG to molecules and surfaces. A number of such derivatives have been prepared, some of which are of somewhat general utility, some of which are directed to solving specific problems associated with particular applications, and some of which have not proved useful or have problems that limit their usefulness. For example, active esters of alpha-, omega-dicarboxylic acids of PEG have been prepared, typically for attachment of PEG to amino groups . An example of such an active ester is poly(ethylene glycol) di - succinimidyl succinate (symbolized in shorthand as "SS- PEG-SS") : H02C-CH2CH2 - C02-PEG-02C- CH2CH2- C02-H + 2NHS -OH → → NHS-02C-CH2CH2 - C02 - PEG-02C- CH2CH2- C02-NHS
SS - PEG-SS
The symbol NHS is shorthand for N-hydroxy succinimide, from which the succinimidyl active ester moiety is derived. In the remaining chemical drawings the N-hydroxylsuccinimide active ester moiety will be represented as -C02-NHS.
The SS-PEG-SS active ester reacts rapidly with amino groups on proteins and other molecules to form a stable amide linkage (-C0-NH-) . However, a problem with the active ester is that SS-PEG-SS possesses ester linkages in the backbone that remain intact after coupling to an amine such as a protein (represented as PRO-NH2) :
SS-PEG-SS + 2PRO-NH2
→ PRO-NHCO-CH2CH2-C02-PEG-02C-CH2CH2-CONH-PRO
The intact ester linkage that remains after the protein and PEG conjugate is formed is subject to hydrolysis. The PEG detaches from the modified protein in aqueous solution. The rate at which hydrolysis occurs can vary with the environment .
U.S. Patent No. 4,670,417 discloses hemoglobin modified with various dicarboxylic acid PEGs in which the ester linkage that would otherwise remain in the conjugate is replaced with an ether linkage. The hemoglobin is modified in the presence of amino acids or amines that are said to also react with the dicarboxylic acid PEG to prevent excessive carboxyl groups from reacting with the hemoglobin, to avoid crosslinking and gelation of the reaction mixture, and to avoid the time, expense, and use of large reaction vessels that would otherwise be required. Thus, special precautions are necessary to prevent cross linking with this difunctional material. PEGs that are activated at each terminus with the same reactive moiety, such as the activated dicarboxylic acid PEGs discussed above, have been referred to in the art as being "homobifunctional" to indicate that the active moieties at the polymer ends are normally the same. There is also much interest in PEG derivatives in which different functional groups are present at the two terminae of the polymer chain. These derivatives are sometimes referred to as heterobifunctional or heterofunctional .
Zalipsky and Barany in J. of Bioactive and Compatible Polymers, 5, 227-231 (1990) describe preparation of the following heterofunctional PEG in which the polymer has a carboxymethyl group at one terminus and a hydroxyl group at the other terminus :
HO-PEG-0-CH2C02-H Both hydroxyl and carboxymethyl groups in the above molecule can be "activated" for subsequent reaction by conversion to other forms. For example, Zalipsky in Bioconiucrate Chemistrv, 4, 296-299 (1993) describes preparation of the following heterofunctional PEG in which the hydroxyl terminus has been converted to the succinimidyl carbonate and the carboxyl terminus has been converted to a complex structure referred to as a t-Boc-protected hydrazide:
NHS-02C-0-PEG-CONH-CH2CO-NH-NH-C02-C (CH3) 3 The above derivative is proposed for attaching amino-phosolipids to the succinimidyl carbonate, followed by removal of the t-Boc protecting group, to give PEG-phospholipids with a reactive hydrazide group remaining at the PEG terminus.
PEG derivatives active on one end and capped on the opposite end by a relatively nonreactive methoxy group have been prepared. These mPEGs are typically considered to be monofunctional, so that only one end of the PEG is active for conjugation to other substances. For example, one PEG is frequently used in a form in which there is a relatively nonreactive methoxy group at one terminus and the reactive succinimidyl succinate group at the other terminus :
CH3O-PEG-02C-CH2CH2-CO2-NHS It is desirable in the biotechnical arts to continually develop activated polymers suitable for conjugation with one or more of various substances including proteins, lipids, liposomes, drugs, surfaces, and ligands for two-phase partitioning. With the foregoing in mind, it is an object of the invention described below to provide additional such activated PEGs.
Summary of the Invention The invention is based upon the recognition that many of the activated carboxylic acid PEGs have poor reactivity and are either too slow or too fast to be generally useful for conjugation. For example, the NHS ester of carboxymethylated PEG is so reactive that it hydrolyzes almost immediately upon solution in water. This high reactivity is a serious deficiency of PEG derivatives based on carboxymethylated PEG.
The invention provides heterofunctional and monofunctional PEGs and related polymers having a single carboxylic acid moiety of suitable reactivity, which can be activated. The active esters of these polymer acids have a single active ester and no other ester linkages. The active esters have a half life in water of from about 10 to 25 minutes. These esters include either a propionic or butanoic acid moiety and a polymer such as PEG or a related polymer. These related polymers include poly(alkylene oxides) , poly(oxyethylated polyols), poly(olefinic alcohols) , and poly(acrylomorpholine) .
Also provided in accordance with the invention are conjugates of these activated polymers with biologically active substances such as polypeptides, proteins, enzymes, phospholipids, lipids, liposomes, nucleosides, oligonucleotides, drugs, dyes, and the surfaces of solid materials that are compatible with living organisms, tissue, or fluids, which are sometimes referred to as biomaterials. Further provided are methods for preparation of these activated polymers and conjugates.
In one embodiment, one terminus of PEG is the activated propionic acid or butanoic acid moiety and the other terminus can be capped with a relatively nonreactive methoxy group, for example, or can be activated with any other functional group, which may be protected or unprotected. For example, the succinimidyl ester of heterofunctional PEG propionic acid can be represented as follows :
X-PEG-0-CH2CH2-C02-NHS
In the above example, "X-" can include any activating group other than a carboxylic acid or activated carboxylic acid or can be a relatively nonreactive group such as methoxy, CH30- . For example, "X-" can include the thiol-selective vinyl sulfone and other sulfone moieties as shown in commonly assigned U.S. Patent No. 5,446,090, which issued on August 29, 1995, the contents of which are incorporated herein by reference. The utility of the heterofunctional and monofunctional propionic and butanoic acid PEGs is believed to come from four sources. First, the succinimidyl ester of PEG propionic acid has ideal reactivity for attachment to amines, such as proteins, in aqueous solution. The succinimidyl ester of PEG butanoic acid is less reactive than the propionic acid, but is still of utility. Second, the reactions at the terminae of the heterofunctional and monofunctional PEG molecules can be controlled to limit cross linking generally more easily than typically is true of homobifunctional derivatives. Third, the hydrolytic stability of the ether linkages in the backbone of PEG propionic acid results in stable conjugates after chemical coupling to another molecule or surface. Fourth, the hydrolytic stability of the ether linkage in the intermediate heterofunctional and monofunctional propionic acid PEGs permits ready purification of the intermediate by ion exchange chromatography in aqueous medium. The structure of the active esters of poly(ethylene glycol) of the invention can be represented as follows :
R- (0CH2CH2)n-Z- (CH2)m-C02-Q The moiety (OCH2CH2)n represents PEG wherein n is from about 20 to 4000. More typically, n is from 20 to
2000. Z is selected from hydrolytically stable groups including -O- , -S-, -NHCO-, -CONH-,
-NHC02-, and -02CNH- . The value of m is from 2 to 3 for propionic and butanoic acid derivatives, respectively. Q is selected from the group including hydrogen, tert- butyl, N-succinimide, N-sulfosuccinimide, N-phthalimide, N-glutarimide, N-tetrahydrophthalimide, N-norbornene-2, 3-dicarboximide, hydroxybenzotriazole, and hydroxy-7-azabenzotriazole. R is selected from the group including hydrogen, aldehyde, acetal, alkyl, benzyl, aryl, alkenyl CH2=CH-CH2-, acrylate CH2=CH-CO-, methacrylate CH2=C(CH3) -CO- , acrylamide CH2=CH-CONH- CH2CH2-, active sulfone, amine H2N-CH2CH2-, and protected amine Y-NH-CH2CH2- (where Y is any of several amine protecting groups well known in the art and including, for example, t-Boc) , thiol HS-CH2CH2-, and protected thiol B-S-CH2CH2- (where B is any of several thiol protecting groups well known in the art and including, for example, orthopyridyldisulfide) . Thus, the invention provides active esters that have suitable reactivity for modification of other molecules and surfaces. Use of these active esters to prepare PEG-proteins, PEG- surf aces, and PEG- phospholipids is described.
Detailed Description of the Invention The following examples are given to illustrate the invention, but should not be considered in limitation of the invention:
Example 1 - Synthesis of CH30- (CH2CH20) n-CH2CH2C02-NHS Example 2 - Synthesis of CH30- (CH2CH20) n-CH2CH2-S- CH2CH2C02-NHS Example 3 - Synthesis of CH30- (CH,CH20) n-CH2CH2CH2C02-NHS Example 4 - Rates of hydrolysis of PEG-NHS active esters Example 5 - Synthesis of HO- (CH-,CH20) n-CH2CH2-S-CH2CH2 -C02H Example 6 - Synthesis of HO- (CH2CH20) n-CH2CH2-S-CH2CH2
-C02-NHS Example 7 - Synthesis CH2=CH-C02 (CH2CH20) nCH2CH2-S
-CH2CH2C02-NHS Example 8 - Synthesis CH2 = CHCH20- (CH2CH20) nCH2CH2S -CH2CH2C02NHS
Example 9 - Synthesis of HO- (CH,CH-,0) n-CH2CH2NHCOCH2
CH2C02C(CH3)3 Example 10 - Synthesis of H2N- (CH2CH20) πCH2CH2-S-CH2CH2C02H Example 11 - Synthesis of HO- (CH2CH20) n-CH2CH2C02H Example 12 - Synthesis of H2N-CH?CH20 (CH2CH20) n-CH2CH2C02H Example 13 - Synthesis of (CH3) -C-O-CONH- (CH2CH20) n-S
-CH2CH2C00H Example 14 - Synthesis of (CH3) -C-O-CONH- (CH2CH20) n -CH2CH2C00H Example 15 - α-methoxy-ω-dipalmitoylphos- phatidylethanolamide of propionic acid of PEG (a"PEG-phospholipid) Example 16 - α-methoxy-ω-distearoyl- phosphatidylethanolamide of propionic acid of PEG (a "PEG"-phospholipid)
Example 17 - Synthesis of HS-CH2CH2CONH- (CH2CH20)n-CH2CH2 -S-CH2CH2C02H Example 18 - Coupling of methoxy-PEG-SPA to glass surfaces Example 19 - Coupling of PEG-SPA to proteins
EXAMPLE 1 alpha-methoxy, omega-propionic acid succinimidyl ester of PEG ("methoxy-PEG-SPA") CH30- (CH2CH20)n-CH2CH2C02-NHS
A mixture of methoxy-PEG (or M-PEG-OH) of molecular weight 5000 (25.0 g) , distilled water (25.0 ml) and potassium hydroxide (0.5 g) was cooled to 0-5°C in an ice bath. Acetonitrile (3.4 g) was added slowly, and the solution was stirred for 2.5 hours at 0-5°C. The pH of the solution was adjusted to 7 by addition of sodium phosphate. The product was extracted with dichloromethane (200, 70 and 50 ml) . The organic layer was dried over magnesium sulfate, diethylether. The precipitate was removed by filtration and dried under vacuum. Yield of PEG nitrile 23.5 g. NMR (d6-DMS0) : 2.74 ppm (t, 4H, -CH2-CN) ; 3.21 ppm, (s, -0CH3) , 3.50 ppm (s, -0CH2CH20-) .
A mixture of M-PEG nitrile from the above step (23.5 g) and concentrated hydrochloric acid (117.5 g) was stirred at room temperature for 48 hours. The solution was diluted with one liter of water and extracted with dichloromethane (200, 150, and 100 ml) . The combined organic extracts were washed twice with water, dried over sodium sulfate, filtered, and concentrated to dryness by rotary evaporation. Yield of PEG amide 21.5 g. NMR (d6-DMSO) : 2.26 ppm (t, 4H, -CH2-C0NH2) ; 2.43 ppm (t, 4H, -CH2-COOH) ; 3.21 ppm (s, -0CH3) , 3.50 ppm (s, -OCH2CH20-) .
M-PEG amide from the above step (16.0 g) was dissolved in 1150 ml of distilled water, 100 g of potassium hydroxide was added, and the solution was stirred for 22 hours at room temperature. Sodium chloride (150 g) was added, and the solution was extracted with dichloromethane (150 ml x 3) . The combined organic extracts were washed with 5% oxalic acid, water (twice) , and dried over sodium sulfate. The solution was concentrated and the product precipitated by addition to diethylether. The product M-PEG propionic acid was collected by filtration and dried over vacuum. Yield of acid 14.0 g. NMR (dg- DMSO) : 2.43 ppm (t, 4H, -CH2-COOH) ; 3.21 ppm (s, -OCH3) , 3.50 ppm (s, -OCH2CH20-) .
M-PEG propionic acid (3.4 g, 1 mmol) was dissolved in dichloromethane (20 ml) and N- hydroxysuccinimide (2.1 mmol) was added. The solution was cooled to 0°C, a solution of dicyclohexylcarbodiimide (2.1 mmol) in 4 ml dichloromethane was added dropwise, and the solution was stirred at room temperature overnight . The reaction mixture was filtered, concentrated, and precipitated by addition to diethylether. Yield of final product: 3.3 g. NMR (d6-DMSO) : 2.81 ppm (s, 8H, NHS); 2.92 ppm (t, 4H, -CH2-COO-) ; 3.21 ppm, (s, -OCH3) , 3.5 ppm (s, -OCH2CH20-) .
EXAMPLE 2 alpha-methoxy, omega-thiopropionic acid succinimidyl ester of PEG
CH30- (CH2CH20)n-CH2CH2S-CH2CH2C02-NHS
In a round bottom flask equipped with a magnetic stir bar, reflex condenser and nitrogen line, M-PEG-5000-methanesulfonate (from Shearwater Polymers, 70.0 g; 0.0206 moles) was added to a mixture of 280 ml toluene and 420 ml absolute ethanol and stirred to dissolve. Sodium hydroxide (3x excess; 2.3 g; 0.058 moles) was dissolved in 56 ml absolute ethanol and added to the PEG-mesylate. Ethyl-3-mercaptopropionoate (3x excess; 7.84 ml; 8.15 g; 0.061 moles) was added to -li¬ the reaction via syringe and the reaction was heated under a nitrogen atmosphere to 60°C for three hours. The reaction mixture was cooled to room temperature, filtered to remove insoluble salts and concentrated to about 250 ml under reduced pressure. This is added to 1200 ml cold diethyl ether, giving a pale yellow precipitate which is dried in vacuo overnight . Yield 70.0 g, NMR (DMSO-d6) 2.63 ppm (m, SCH2CH2, 4H) , 3.21 ppm, (s, -OCH3) , 3.50 ppm (s, PEG backbone, 304 H) , 4.57 ppm (t, OH, IH) .
In an Erlenmeyer flask equipped with a magnetic stir bar, M-PEG-5000-S-CH2CH2-COOC2HS from the preceding step (70.0 g; 0.0206 moles) was added to 840 ml distilled deionized water and stirred to dissolve. In a separate beaker, sodium hydroxide (1.4 g) was added to 35 ml distilled deionized water and stirred to dissolve. The sodium hydroxide solution was added to the PEG solution until the pH was 12-13. The solution was then stirred at room temperature for one hour. Oxalic acid was added to adjust the pH to 3. The solution was extracted with CH2C12 three times (200/200/200 ml) . The extract was dried over Na2S04/MgS04, filtered, concentrated under reduced pressure to about 150 ml and added to 1000 ml cold diethyl ether to precipitate the product. The product was dried in vacuo overnight. Yield: 61.6 g, 88%. 1HNMR(DMSO-d6) 2.625 ppm (m, CH2CH2, 4H) , 3.21 ppm (s, - OCH3) , 3.50 ppm (s, PEG backbone, 304 H) .
In a round bottom flask equipped with a magnetic stir bar and a rubber septum attached to a nitrogen line and bubbler, M-PEG-5000-COOH from the preceding step (3.00 g; 0.8823 mmol) , N,N' dicyclohexyl carbodiimide (1.5x excess; 0.2731 g; 0.001324 mmol) and N-hydroxysuccinimide (1.5x excess; 0.1523 g; 0.001324 mmol) were dissolved in 60.0 ml CH2C12. The flask was immersed in an ice bath and stirred overnight. The reaction mixture was filtered, concentrated under reduced pressure, filtered and precipitated into cold diethyl ether. Yield 2.50 g, 83%. 1HNMR (DMSO-d6) 2.625 ppm (m, CH2CH2, 4H) , 2.81 (s, succinimide, 4H) , 3.21 ppm, (s, -OCH3) , 3.50 ppm (s, PEG backbone, 304 H) .
EXAMPLE 3 alpha-methoxy, omega-butanoic acid succinimidyl ester of PEG
CH30- ( CH2CH20 ) n- CH2CH2CH2C02 -NHS
Ethyl malonate (3.5 ml, 2 equivalents) dissolved in 100 ml of dioxane were added drop by drop to sodium hydride (0.674 g, 2 equivalents) and toluene
(50 ml) in a round bottomed flask under nitrogen flow.
M-PEG methanesulfonate (20 g, 1 equivalent, MW 5000 daltons, from Shearwater Polymers) was dissolved in 50 ml of toluene and added to the above mixture. The resulting mixture was reflexed overnight. The reaction mixture was then concentrated to half its original volume, extracted with 15 ml of 10% aqueous NaCl solution, extracted with 10 ml of 1% aqueous hydrochloric acid, and the aqueous extracts combined. The collected aqueous layers were extracted with dichloromethane (50 ml x 3) , and the organic layer was dried with magnesium sulfate for 3 hours, filtered, and evaporated to dryness. Yield: 20 g of PEG malonic ester. NMR (d6-DMS0) : 1.17 ppm (t, 12H, -CH3) ; 1.99 ppm (quartet, 4H, -CH2-CH) ; 3.21 ppm, (s, -0CH3) ; 3.5 ppm (s, -0CH2CH20-) ; 4.10 ppm (quintet, 8H, -OCH-,-CHJ .
M-PEG malonic ester (18 g) was dissolved in 240 ml of IN sodium hydroxide containing 12 g of sodium chloride, and the mixture was stirred for one hour.
The pH of the mixture was adjusted to 3.0 by addition of 6N hydrochloric acid, and the mixture was extracted with dichloromethane (150 ml and 100 ml) . The organic layer was dried over magnesium sulfate, filtered, concentrated, and poured into cold diethylether. The product M-PEG malonic acid was removed by filtration and dried under vacuum. Yield: 16 g. NMR (d6-DMS0) ; l.o ppm (q, 4H, -CH2CH2CH) ; 2.90 ppm (t, 2H, -CH2CH-) ; 3.21 ppm (s, -OCH3) ; 3.5 ppm (s, -OCH2CH20-); 12.1 ppm (s, 2H, -COOH) .
M-PEG malonic acid (15 g) was dissolved in 120 ml of dioxane and refluxed for 8 hours, then concentrated to dryness. The residue was dissolved in 100 ml water, extracted with dichloromethane (70 ml and 50 ml) , dried over magnesium sulfate, and the solution concentrated by rotary evaporation. The residue was precipitated by addition to cold diethyl ether. Yield: 11 g. NMR (d6-DMSO) : 1.72 ppm (quintet, 4H, -CH2CH2CH2- COOH) ; 2.4 ppm (t, 4H, -CH2CH2CH2-COOH) ; 3.21 ppm (s, - OCH3) ; 3.37 ppm (t, 4H, -CH2CH2CH2-COOH) ; 3.5 ppm (s, - OCH2CH20-) .
M-PEG butanoic acid (3.4 g, 1 mmol) was dissolved in dichloromethane (20 ml) and N- hydroxysuccinimide (2.1 mmol) was added. The solution was cooled at 0°C, a solution of dicyclohexylcarbodiimide (2.1 mmol) in 4 ml dichloromethane was added dropwise, and the solution was stirred at room temperature overnight . The reaction mixture was filtered, concentrated, and precipitated by addition to ethyl ether. Yield of final product: 3.3 g. NMR (d6-DMSO) : 1.83 ppm (quintet, 4H, -CH,CH,CH,-COO-) ; 2.70 ppm (t, 4H, -CH2-COO-) ; 2.81 ppm (8H, NHS) ; 3.21 ppm (s, -OCH3) ; 3.5 ppm (s, - OCH2CH20-) .
EXAMPLE 4
RATES OF HYDROLYSIS OF PEG-NHS ACTIVE ESTERS
To determine the relative reactivities of the various PEG-NHS active esters, rates of hydrolysis were determined by using UV spectroscopy to monitor the formation of N-hydroxylsuccinimide at 290 nm. In a typical experiment, 7-10 mg of PEG-NHS ester was dissolved in 3.0 ml of 0.1 M phosphate buffer, pH 8.0, and the increase in absorbance at 290 nm was followed. A plot of log of absorbance at time zero minus absorbance at time t versus time yields a straight line of slope equal to the first order rate constant for hydrolysis. Division of this rate constant into 0.693 gives the half life for hydrolysis. All rates were performed in duplicate. Table 1 presents the half lives for hydrolysis of a series of PEG-NHS esters, including the new compounds prepared in this work as well as some known compounds. Additional experiments have shown that aminolysis rates parallel these hydrolysis rates.
Table 1. Hydrolysis half lives at pH 8 !, 25 °C, for PEG-succinimidyl esters half life
PEG NHS Ester Symbol (minutes)b
PEG-O-CH2CH2CH2-CO2-NHS (1, SBA) 23.3
PEG-O-CO2-NHS (2, SC) 20.4
PEG-O2C-CH2CH2CH2-CO2-NHS (3, SG) 17.6
PEG-O-CH2CH2-CO2-NHS (4, SPA) 16.5
PEG-S-CH2CH2-CO2-NHS (5, SSPA) 10.7
PEG-02C-CH2CH2-CO2-NHS (6, SS) 9.8
PEG-O2CNH-CH(Bu)-CO2-NHS (7, NOR) 5.4
PEG-O2CNH-CH(R)-CO2-NHSa (8, PEG2) 4.9
PEG-NHCO-CH2CH2-CO2-NHS (9, SSA) 3.2
PEG-O-CH2-CO2-NHS (10, SCM) 0.75
\ R = CH2CH2CH2CH2-CO2-NHCO2 -PEG. b. As a rough rule of thumb the half 1 ife triples upon lower ing pH one unit.
As shown in the table, SCM-PEG (10) has a half life of 0.75 minutes and reacts with water too quickly to be useful for conjugation with biologically active substances or surfaces. SS-PEG (6) , with a half life of 9.8 minutes, has good reactivity, but contains an ester linkage and forms hydrolytically unstable conjugates. On the other hand, SSPA-PEG (5), which is prepared in accordance with the present invention, has good reactivity and no ester linkage that could interfere with hydrolytic stability of conjugates.
SPA-PEG (4) has ideal reactivity at a half life of 16.5 minutes and no ester linkage that could interfere with hydrolytic stability. SBA-PEG (1) reacts more slowly, at a half life of 23.3 minutes, but still is of utility. Generally, a half life of 10 to about 20 or 25 minutes is desirable.
EXAMPLE 5
Synthesis of α-hydroxyl-ω-carboxyl-PEG
HO- ( CH2CH20) n- CH2CH2 - S -CH2CH2- C02H
In a round bottom flask equipped with a magnetic stir bar, reflex condenser and nitrogen line, PEG-3400-methanesulfonate (70.0 g; 11% substituted; 0.0206 moles, Shearwater Polymers) was added to a mixture of 280 ml toluene and 420 ml absolute ethanol and stirred to dissolve. Sodium hydroxide (3x excess; 2.32 g; 0.0581 moles) was dissolved in 56 ml absolute ethanol and added to the PEG-mesylate. Ethyl-3- mercaptopropionoate (3x excess; 7.84 ml; 8.15 g; 0.0607 moles) was added to the reaction via syringe and the reaction was heated under a nitrogen atmosphere to 60°C for three hours. The reaction mixture is cooled to room temperature, filtered to remove insoluble salts and concentrated to about 250 ml under reduced pressure. This is added to 1200 ml cold diethyl ether, giving the desired ester as a pale yellow precipitate which is dried in vacuo overnight .
In an Erlenmeyer flask equipped with a magnetic stir bar, PEG ester from the preceding step (70.0 g; 0.0206 moles) was added to 840 ml distilled deionized water and stirred to dissolve. In a separate beaker, sodium hydroxide (1.4 g) was added to 35 ml distilled deionized water and stirred to dissolve. The sodium hydroxide solution was added to the PEG solution until the pH was 12-13. The solution was then stirred at room temperature for one hour. Oxalic acid was added to adjust the pH to 3. The solution was extracted with CH2C12 three times (200/200/200 ml) . The extract was dried over Na2S04/MgS04, filtered, concentrated under reduced pressure to about 150 ml and added to 1000 ml cold diethyl ether to precipitate the product . The product was dried in vacuo overnight . Yield: 61.6 g, 88%. The mixture was purified by ion exchange chromatography on Sepharose FF (Pharmacia) . -NMR (DMSO-d 2.63 ppm (m, -SCH2CH2-, 4H) , 3.50 ppm (s, PEG backbone, 304H) , 4.57 ppm (t, OH, IH) .
EXAMPLE 6 Synthesis of α-hydroxy-ω-succinimidyl-PEG HO- (CH2CH20)n-CH2CH2-S-CH2CH2-C02-NHS
In a round bottom flask equipped with a magnetic stir bar and a rubber septum, attached to a nitrogen line and a bubbler, hydroxyl-acid from the preceding step (3.00 g; 0.882 mmol) , N,N' dicyclohexyl carbodiimide (1.5x excess; 0.273 g; 0.00132 mmol) and N-hydroxysuccinimide (1.5x; 0.152 g; 0.00132 mmol) were dissolved in 60 ml CH2C12. The flask was immersed in an ice bath and stirred overnight. The next day the reaction mixture was filtered, concentrated under reduced pressure, filtered and precipitated into cold diethyl ether. Yield 2.50 g, 83%. "HNMR(DMSO-d6) 2.625 ppm (m, CH2CH2,4H) , 2.81 ppm, (s, succinimide, 4H) , 3.50 ppm (s, PEG backbone, 304 H) , 4.57 ppm (t, OH, IH) . EXAMPLE 7 Synthesis of ω-succinimidyl-α-acryloyl PEG
CH2=CH-C02- (CH2CH20)nCH2CH2S-CH2CH2-C02-NHS
In a round bottom flask equipped with a mechanical stir bar and a rubber septum attached to a nitrogen line and a bubbler, ω-carboxyl -a-hydroxyl-PEG from Example 5 (1.00 g; 0.000294 moles) was dissolved in 30.0 ml dry CH2C12. Triethylamine (2.5 x excess; 0.0744 g; 0.00103 moles) was weighed in a small test tube, diluted with 1 ml CH2C12 and added via syringe.
Acryloyi chloride, 98% (2.5 x excess; 0.0665 g; 0.00103 moles) was weighed in a small test tube, diluted with 1 ml CH2C12 and added via syringe. The reaction was stirred under nitrogen overnight. The mixture was concentrated under reduced pressure until slightly viscous and dripped into 80 ml cold diethyl ether to precipitate the product . After drying in vacuo three hours, the product was dissolved in distilled deionized water (100 ml) and stirred for 30 minutes. The aqueous solution was extracted in CH2C12 three times (30/30/20 ml) . The extract was dried over Na2S04/MgS04, concentrated under reduced pressure until slightly viscous, and dripped into 100 ml cold diethylether to precipitate the product. Yield: 0.946 g, 95%. Hl-NMR (DMS0-d6) 2.625 ppm (m, SCH,CH,, 4H) , 3.50 ppm (s, PEG backbone, 304 H) , 5.96 ppm (dd, CHaHbCHcCOO, IH) , 6.20 ppm (dd, CHaHbCHcCOO, IH) , 6.35 pm (dd, CHaHbCHcCOO. IH) .
In a round bottom flask equipped with a magnetic stir bar and a rubber septum attached to a nitrogen line and a bubbler, this product (0.946 g; 0.278 mmol), N,N' dicyclohexyl carbodiimide (1.5x excess; 0.0861 g; 0.417 mmol) and N-hydroxysuccinimide (1.5x excess; 0.0480 g; 0.417 mmol) were dissolved in 30.0 ml CH2C12. The flask was immersed in an ice bath and stirred overnight. The next day the reaction mixture was filtered, concentrated under reduced pressure, filtered and precipitated into cold diethyl ether. Yield 0.592 g., 63%. αH-NMR (DMSO- 6) : 2.625 ppm (m, SCH2CH2, 4H) , 2.81 ppm, (s, succinimide, 4H) , 3.50 ppm (s, PEG backbone, 304 H) , 5.96 ppm (dd,
CHaHbCHcCOO, IH) , 6.20 ppm (dd, CHaHbCHcCOO, IH) , 6.35 ppm (dd, CHaHbCHcCOO, IH) . Hydrolysis, followed by titration of the resulting acid, indicated 2.49 x IO"4 moles of acid per gram of PEG, or 85% of the theoretical value. FT-IR: 1783 cm"1 C=0 stretch, (succinimidyl ester end groups) , 1114 cm"1 C-O-C stretch.
EXAMPLE 8 Synthesis of α-allyl-ω-mercaptopropionic acid-PEG CH2=CH-CH2-0- (CH2CH20)nCH2CH2-S-CH2CH2-CO2-NHS
In a three neck 1000 ml round bottom flask equipped with a magnetic stir bar, a Dean-Stark trap, a reflux condenser and a drying tube filled with CaCl2, PEG 3400 (60.0 g; 0.0176 moles) was dissolved in 500 ml toluene. The mixture was azeotropically dried at 130 degrees C for approximately 20 minutes, such that 15-20 ml cloudy solution (solvent/water) collected in the Dean-Stark trap. After draining the Dean-Stark trap, the mixture was cooled to room temperature under nitrogen and 25% CH3ONa in methanol (4.03 ml; 0.0174 moles) was added via syringe. The reaction was heated to reflux and the methanol was collected in the Dean- Stark trap along with an additional 8 ml solvent. The reaction mixture was cooled under nitrogen to room temperature, during which time the color changed from pale yellow to deep red-brown. Allyl chloride (1.35 g; 1.43 ml; 0.00176 moles) was added via syringe and the mixture was stirred overnight at room temperature. The following morning the mixture was filtered through a buchner funnel, concentrated under reduced pressure and precipitated into cold ether. Yield 49.17 g, 82%. The isolated product was checked by NMR to confirm that substitution had occurred. In a 500 ml two neck round bottom flask equipped with a magnetic stir bar, Dean-Stark trap, reflux condenser and drying tube filled with CaCl2, allyl-PEG 3400 (49.17 g; 0.0144 moles) was dissolved in 250 ml toluene and azeotropically dried approximately 15 minutes to remove 15 ml cloudy solvent/water. After cooling under nitrogen to room temperature, distilled triethylamine stored over KOH (3x excess, 4.39 g; 6.05 ml; 0.0433 moles) was added via syringe followed by the addition of distilled methanesulfonyl chloride (3x excess; 4.97 g; 3.36 ml) and 35 ml dry CH2C12. The mixture was stirred at room temperature under nitrogen overnight. The next morning the mixture was concentrated under reduced pressure to remove the CH2C12, filtered through a glass frit and concentrated under reduced pressure to approximately 100 ml. The reaction mixture was placed in a 500 ml round bottom flask equipped with a magnetic stir bar and a reflux condenser fitted with a rubber septum. A small amount was isolated to confirm complete substitution by NMR. Three hundred ml absolute ethanol were added to the reaction mixture. Sodium hydroxide (4x excess; 2.77 g; 0.0692 moles) was dissolved in 60 ml absolute ethanol and added to the reaction mixture. Ethyl-3-mercaptopropionoate (4x excess; 9.31 g; 0.069 moles) was added to the flask via syringe and the reaction mixture was heated to 60°C under nitrogen for three hours. The reaction mixture was then cooled to room temperature, filtered to remove insoluble salts, and concentrated under reduced pressure to approximately 150 ml. The viscous yellow syrup was dripped into 600 ml cold diethyl ether to precipitate a pale yellow product. Yield 44.58 g, 91%. Esterification was confirmed by NMR. After overnight drying under high vacuum, the product was placed in a 1000 ml Erlenmeyer flask equipped with a magnetic stir bar and dissolved in 550 ml distilled deionized water. Using a pH meter, 1.0M NaOH was added until the pH remained stable at 12. This process took approximately 1 1/2 hours. The mixture was then acidified to pH 3.0 with oxalic acid, and extracted with CH2C12 (150/100/100) The extract was dried with Na2S04/MgS04, concentrated under reduced pressure and dripped into 500 ml cold diethyl ether to precipitate the product. Yield 31.55 g, 71% XHNMR (DMSO-dJ 2.625 ppm (m, SCH2CH2, 4H) , 3.50 ppm (s, PEG backbone, 304 H) , 3.95 ppm (dd,CH2=CHCH20, 2H) , J = 13 Hz, 5.13 ppm (dd) , CHaHb=CHcCH,Q, IH) , J = 13 Hz, 5.24 ppm (dd, CHaHb=CHcCH,0, IH) , J= 18 Hz, 5.88 ppm (dd, CHaHb=CHcCH20, IH) . FT-IR data: v 2879 cm-1, C-H stretch (PEG) .
In a 100 ml round bottom flask equipped with a magnetic stir bar, a rubber septum attached to a nitrogen line and a bubbler, α-allyl-ω- mercaptopropionic acid-PEG 3400 (2.052 g; 0.604 mmol), N,N' dicyclohexylcarbodiimide (1.5 x excess, 0.187 g; 0.905 mmol) and N-hydroxysuccinimide (1.5 x excess, 0.104 g; 0.905 mmol) were dissolved in 40 ml dry CH2C12. The flask was immersed in an ice bath and stirred overnight. The following day the reaction mixture was filtered, concentrated under reduced pressure, filtered again and precipitated into 60 ml cold diethyl ether. Yield 1.075 g, 52% 'HNMR (DMSO-dJ 2.625 ppm (m, SCH2CH2, 4H) , 2.81 ppm (s, succinimide, 4H) , 3.50 ppm (s, PEG backbone, 304 H) , 3.95 ppm (dd, CH2CHCH20, 2H) , J = 13
Hz, 5.13 ppm (dd, CHaHbCHcCH20, IH) , J = 13 Hz, 5.24 ppm (dd, CHaHbCHcCH20, IH) , J" = 18 Hz, 5.88 ppm (dd, CHaHbCHcCH20, IH) . Chromatography gave an absorbance at 7.10 ml (14.2 min.) corresponding to the mono acid and a negative absorbance at 7.95 ml (15.9 min.) corresponding to a change in salt concentration for the buffer. An absorbance at 8.20 ml, (16.4 min.) corresponded to hydrolyzed N-hydroxysuccinimidyl ester. Titration data indicates, 2.69 x 10"4 moles/gram PEG, 87.7% of the theoretical value. FT-IR data; v 2879 cm"1, C-H stretch (PEG) .
EXAMPLE 9 alpha -hydroxy, omega- tert -butyl ester of carboxyl PEG HO- ( CH2CH20 ) n- CH2CH2NHCOCH2CH2COOC ( CH3 ) 3
A mixture of α-hydroxy-ω-amine of PEG of molecular weight 3400 (from Shearwater Polymers, 17 g, 0.00050 moles) , tert-butyl hydrogen succinate (4.35 g, 0.025 moles, 5 fold molar excess) , N,N-dicyclohexyl carbodiimide (5.16 g, 0.025 moles, 5 fold molar excess) , and dichloromethane (100 ml) was stirred 16 hours at room temperature under nitrogen atmosphere. The mixture was filtered and solvent was distilled off under reduced pressure. The solid residue after distillation was dissolved in 400 ml distilled water and stirred overnight. Next the solution was filtered and the reaction product was extracted with dichloromethane (3 X 50 ml) . The extract was dried with anhydrous sodium sulfate and added to 800 ml cold diethyl ether. The precipitated product was filtered off and dried under reduced pressure. Yield 15.8 g, 88.9% JH NMR (DMSO-d6) : 1.37 ppm (s, (CH3)3C, 9H) , 2.33 ppm (m, -OOC-CH2CH2-COO- , 4H) , 3.50 ppm (s, PEG backbone, 304 H) , 4.57 ppm (t, OH, IH) .
EXAMPLE 10 alpha-amino, omega-thiopropionic acid of PEG
H2NCH2CH20 ( CH2CH20) nCH2CH: - S - CH2CH2C02H
PEG- 3350 -methanesufonate ( from Shearwater
Polymers 100 . 0 g ; 0 . 0281 moles ) was added to a mixture of 400 ml toluene and 300 ml absolute ethanol and stirred to dissolve . Ethyl - 3 -mercaptopropionate ( 6 . 0 g; 0.0445 moles. 79.2% of stoichiometric amount) and sodium hydroxide (1.6 g; 0.04 moles, 71.1% of stoichiometric amount) dissolved in 40 ml absolute ethanol were added to the reaction mixture. The reaction was heated under nitrogen atmosphere at 60°C for three hours. The reaction mixture was cooled to room temperature, filtered to remove insoluble salt and concentrated to about 300 ml under reduced pressure. This is added to 1000 ml cold diethyl ether giving the white precipitate, which was dried in vacuo overnight. The dried white precipitate was added to 180 ml distilled water and stirred to dissolve. The 1.0 M sodium hydroxide solution was added to above solution to adjust pH to 12. The solution was stirred at room temperature for two hours . The pH of the solution was maintained with periodical dropwise addition of 0.1 M sodium hydroxide. Next the solution was added to the mixture of 600 ml concentrated ammonium hydroxide and 60 g ammonium chloride and stirred 44 hours at room temperature. The reaction product (mixture of a , ω- diamino of PEG, α-amino-ω-thiopropionic acid of PEG, and α,ω-dithiopropionic acid of PEG) was extracted with dichloromethane (3 X 300 ml) . The extract was dried over anhydrous magnesium sulfate, filtered and concentrated to dryness. Pure α-amino-hydrochloride-ω- thiopropionic acid of PEG was separated by ion exchange chromatography on S-Sepharose FF (Pharmacia) . Yield 37.5 g. Η NMR (DMSO-d6) : 2.67 ppm (m, -CH2-S-CH2-, 4H) , 2.96 ppm (t, -CH,NH,HC1, 2H) , 3.50 ppm (s, PEG backbone, 304H) .
EXAMPLE 11 alpha-hydroxy, omega-propionic acid of PEG
HO- (CH2CH20)n-CH2CH2C02H
A mixture of PEG of molecular weight 3350 (20.0 g; 0.00597 moles) , distilled water (20.0 g) and potassium hydroxide (0.4 g) was cooled to 0.5°C in an ice bath. Acrylonitrile (0.5 g; 0.00942 moles) was added slowly, and the solution was stirred 2 hours at 0-5°C. Concentrated sulfuric acid (30.0 g) was added and the mixture was heated 3 hours at 95-100°C. After cooling to room temperature distilled water (200 ml) was added, and the reaction product (mixture of unreacted PEG, α-hydroxy-ω-propionic acid of PEG, and dipropionic acid of PEG) was extracted with dichloromethane (3X30 ml) . The extract was dried with anhydrous sodium sulfate, and solvent was distilled off under reduced pressure. Pure α-hydroxy-ω-propionic acid of PEG was separated by ion exchange chromatography on DEAE Sepharose FF (Pharmacia) . Yield 7.2 g. XH NMR (DMSO-d6) : 2.43 ppm (t, -CHo-COOH. 2H) , 3.50 ppm (s, PEG backbone, 304 H) , 4.57 ppm (t, OH, IH) .
EXAMPLE 12 alpha-amino, omega-propionic acid of PEG H2NCH2CH20 ( CH2CH20 ) nCH2CH2C02H
A mixture of α-hydroxy-ω-propionic acid PEG of molecular weight 3400 from Example 11 (8.0 g, 0.00233 moles) , methyl alcohol (90.0 ml) , and concentrated sulfuric acid (0.55 ml) was stirred 1 hour at room temperature. Distilled water (50.0 ml) was added and calcium carbonate was used to adjust the pH of the mixture to 7. Methyl alcohol was distilled off under reduced pressure, and the reaction product α- hydroxy-ω-methyl ester of propionic acid of PEG was extracted with dichloromethane (3 X 30 ml) . Next dichloromethane was distilled off under reduced pressure.
To the solution of a -hydroxy-ω-methyl ester of propionic acid of PEG (5.0 g, 0.00145 moles) in dichloromethane (30 ml) triethylamine (0.27 ml, 0.00193 moles, 133 % of stoichiometric amount) and methanesulfonyl chloride (0.12 ml, 0.00155 moles, 107% of stoichiometric amount) were added and the resulting mixture was stirred overnight under nitrogen atmosphere. The mixture was filtered and solvent was distilled off under reduced pressure. The residue after distillation was dissolved in distilled water (50 ml) and IM sodium hydroxide was added to adjust pH to 12. The solution was stirred at room temperature for one hour. The pH of the solution was maintained with periodical addition of 0.1 N sodium hydroxide. Next the solution was added to the mixture of 160 ml concentrated ammonium hydroxide and 16 g ammonium chloride and stirred 46 hours at room temperature. The reaction product was extracted with dichloromethane (3 X 50 ml) . The extract was washed with 20 ml 1 M hydrochloric acid, 20 ml distilled water and dried with anhydrous sodium sulfate. Next the solvent was distilled under reduced pressure giving 4.2 g of ex¬ amine hydrochloride-ω-propionic acid of PEG. 1H NMR (DMSO-d6) : 2.43 ppm (t, -CH;-COOH, 2H) , 2.96 ppm (t, -CH2NH2HC1, 2H) , 3.50 ppm (s, PEG backbone, 304 H) .
EXAMPLE 13 alpha-tert -butyloxycarbonylamino , omega - thiopropionic acid of PEG ( CH3 ) 3COCONH- ( CH2CH20 ) n- S - CH2CH2COOH
A mixture of α-aminohydrochloride-ω- thiopropionic acid of PEG of molecular weight 3470 from Example 10 (17.4 g, 0.0050 moles) , triethylamine (1.52 g, 0.0150 moles) , di-tert-butyl dicarbonate (1.2 g, 0.0055 moles) , and dichloromethane (100 ml) was stirred 16 hours at room temperature under nitrogen atmosphere. The solvent was distilled off under reduced pressure. The residue was dissolved in 200 ml distilled water, the pH of the solution was adjusted with 1 M HCl to 3, and the reaction product was extracted with dichloromethane (3 X 50 ml) . The extract was dried with anhydrous sodium sulfate and added to 80 ml cold diethyl ether. The precipitated white product was filtered off and dried under reduced pressure. Yield 14.3 g, 80.9%. XH NMR (DMSO-d6) : 1.36 ppm (s, (CH3)3C-, 9H) , 2.67 ppm (m, -CH2-S-CH2-, 4H) , 3.15 ppm (q, -NHCH2CHQ- , 2H) , 3.50 ppm (s, PEG backbone, 304 H) , 6.76 ppm (t, -NH- , IH) .
EXAMPLE 14 alpha-1ert-butyloxycarbonylamino, omega-propionic acid of PEG
( CH3 ) 3COCONH- ( CH2CH20 ) nCH2CH2COOH
A mixture of α-amino hydrochloride-ω- propionic acid of PEG of molecular weight 3460 from Example 12 (17.3 g, 0.0050 moles) , triethylamine (1.52 g, 0.0150 moles) , di-tert-butyl dicarbonate (1.2 g. 0.0055 moles) , and dichloromethane (100 ml) was stirred 16 hours at room temperature under nitrogen atmosphere. The solvent was distilled off under reduced pressure. The residue was dissolved in 200 ml distilled water, the pH of the solution was adjusted with 1 M HCl to 3, and the reaction product was extracted with dichloromethane (3 X 50 ml) . The extract was dried with anhydrous sodium sulfate, and added to 800 ml cold diethyl ether. The precipitated white product was filtered off and dried under reduced pressure. Yield 14.1 g. JI NMR (DMSO-d6) : 1.36 ppm (s, (CH3)3C-, 9H) , 2.43 ppm (t, -CH2COO-, 2H) , 3.15 ppm (q, -NHCH2CH20- , 2H) , 3.50 ppm (s, PEG backbone, 304 H) , 6.76 ppm (t, -NH-, IH) . EXAMPLE 15 α-methoxy-ω-dipalmitoylphosphatidylethanolamide of propionic acid of PEG α-Methoxy-ω-propionic acid succinimidyl ester of PEG of molecular weight 5170 from Example 1 (MSPA)
(4.00 g, 0.000773 moles) was dissolved in chloroform
(40 ml) and treated with solid dipalmitoylphosphatidyl- ethanolamine (DPPE) (0.60 g, 0.000867 moles) and triethylamine (0.70 ml) . The suspension was vigorously mixed and maintained at 45°C for 20 min. During this time a clear solution was formed. TLC (chloroform- methanol-water 90:18:2) showed complete disappearance of MSPA (Rf = 0.73 I2 vapor visualized) and formation of a new product (Rf = 0.58, I2 vapor visualized) . Acetic acid (0.20 ml) and acetonitrile (112 ml) were added and the reaction mixture was cooled to 4°C and then filtered and evaporated to dryness. The solid residue was dissolved in water and the pH of the solution was adjusted to 7.0 with 0.1 N sodium hydroxide. The solution was transferred into Spectrapor nr 2 dialysis tubing (MWCO 12,000-14,000) and dialyzed against 50 mM saline solution 12 hours at 4°C, then two times against distilled water. Next the solution was lyophilized producing 3.3 g of white solid product. XH NMR (CDC13) : 0.88 ppm (t, CH3-CH2-, 6H) , 1.26 ppm (s, CH3, -_[CH2112- , 48H) , 1.58 ppm (m, CH2, 4H) , 2.28 ppm (two overlapping t, CH2-C=0, 6H) , 3.36 ppm (m, -0CH,-CH,NH- , 2H) , 3.64 ppm (s, PEG backbone, 304H) , 3.94 ppm (m, -CH2CH20-P, 2H) , 4.17 ppm (dd, glycerol CH20-P, 2H) , 4.39 ppm (dd, glycerol CH2OOC-, 2H) , 5.20 ppm (m, glycerol Ch, IH) , 7.60 ppm (t, NH, IH) .
EXAMPLE 16 α-methoxy-ω-distearoylphosphatidylethanolamide of propionic acid of PEG
α-Methoxy-ω-propionic acid succinimidyl ester of PEG of molecular weight 2170 (MSPA) (10 g, 0.00461 moles) was dissolved in chloroform (40 ml) and treated with solid distearoylphosphatidylethanolamine (DSPE) (3.72 g, 0.00497 moles) and triethylamine (2.4 ml) . The suspension was vigorously mixed and maintained at 45°C for 20 min. During this time a clear solution was formed. TLC (chloroform-methanol-water 90:18:2) showed complete disappearance of MSPA (Rf = 0.73, I2 vapor visualized) and formation of a new product (Rf = 0.51, I2 vapor visualized) . Acetic acid (0.75 ml) and acetonitrile (160 ml) were added and the reaction mixture was cooled to 4°C and then filtered and evaporated to dryness. The solid residue was dissolved in water and the pH of the solution was adjusted to 7.0 with triethylamine. The solution was transferred into Spectrapor dialysis tubing (nr. 2 dialysis tubing)
(MWCO 12,000-14,000) and dialyzed against 50 mM saline solution 12 hours at 4°C, then two times against distilled water. Next the solution was lyophilized producing 9.2 g of white solid product. NMR (CDC13) : 0.88 ppm (t, CH3-CH2-, 6H) , 1.26 ppm (s, CH,- (CHJ,,-, 56H) , 1.58 ppm (m, CH2, 4H) , 2.28 ppm (two overlapping t, CH2-C=0, 6H) , 3.36 ppm (m, -OCH,-CH,NH- , 2H) , 3.64 ppm (s, PEG backbone, 304H) , 3.94 ppm (m, - CH CHO-P, 2H) , 4.17 ppm (dd, glycerol CH20-P, 2H) , 4.39 ppm (dd, glycerol CH2OOC- , 2H) , 5.20 ppm (m, glycerol CH, IH) , 7.60 ppm (t, NH, IH) .
EXAMPLE 17
Synthesis of HS-CH2CH2CONH- (CH2CH20) n- CH2CH2-S-CH2CH2C02H
PEG amino acid (ω-aminohydrochloride-α-S- propionic acid, from Example 10) (1.83 grams) of molecular weight 400 was dissolved in 250 ml of benzene containing 1.0 ml of triethylamine and 20 ml of dry methylene chloride. Benzene was distilled off under reduced pressure. The residue was redissolved in 10 ml of acetonitrile, and succinimidyl-3- (2- pyridyldithio)propionate (SPDP) dissolved in 10 ml of acetonitrile was added. The mixture was stirred at room temperature under nitrogen atmosphere overnight . Gel permeation chromatography showed no aminoacid peak and a new monoacid peak. The solvent was distilled off under reduced pressure. Yield 3.2 g.
The product from the preceding step (3.2 g) was dissolved in 300 ml of distilled water containing 2.5 g of dithiothreitol and stirred at room temperature under nitrogen atmosphere for 3 hours . The reaction mixture was applied to a DEAE Sepharose FF column (100 ml) , and the column was washed with 800 ml of distilled water. The product was eluted with 200 ml of 0.5 M NaCl . The pH of the eluate was adjusted to 3.0 with
0.1 M HCl, and the product was extracted with methylene chloride (3 X 40 ml) . Next the solvent was distilled off. Yield 1.2 g. Gel permeation chromatograph showed 90% of the desired product, 8% dimer and 2% unknown impurity. -NMR (d6-DMSO) : 2.25 ppm (t, -SH, IH) , 2.38 ppm (t, CH2CONH, 2H) , 2.51 ppm (t, CH2COOH, 2H) , 2.62 ppm (broad m, HS-CH2-, -CH2-S-CH2- , 6H) , 3.20 ppm (q, -C0NHCH2-, 2H) , 3.51 ppm (s, polymer backbone, 36H) , 7.97 ppm (t, NH, IH) .
EXAMPLE 18
Coupling of methoxy-PEG-SPA to glass surfaces
Quartz slides were cleaned and activated for surface modification by soaking in 1% (w/w) aqueous NaOH at 90°C for 10 minutes, 3% (w/w) HCl at 90°C for 10 minutes, and boiling in 30% (w/w) H20: for 1 hour to remove trace organics, then rinsed with water.
The clean glass slides were prepared for functionalization with aminopropylsilane by drying under vacuum of IO"3 torr for 1 hour to remove excess surface water. The glass slides were then exposed to a 2% (v/v) solution of silane in anhydrous toluene for 4 hours at room temperature. The capillaries were rinsed with toluene and cured in a vacuum oven at 190°C, IO"3 torr for 12 hours. This procedure gave quartz slides with available amino groups on the surface.
Methoxy-PEG-SPA from Example 1 (MW 5000) was grafted to the functionally activated quartz surfaces by reacting as 5% (w/v) solution in 0.05 M sodium bicarbonate (pH 8.3) for 4 hours at 40°C. After PEG immobilization, surfaces were rinsed with toluene, dried under vacuum and rinsed with water. Examination with X-ray photoelectron spectroscopy (XPS) showed the presence of a large C-0 peak consistent with attachment of PEG. Also the water-contact angle is near zero, as expected for a PEG-coated surface. Finally, adsorption studies with fibrinogen revealed that fibrinogen adsorption on the PEG-coated surface has been reduced by approximately 98% relative to uncoated quartz.
EXAMPLE 19 Coupling of PEG-SPA to proteins
Succinimidyl esters of PEG propionic acids ("PEG-SPA") derivatives couple readily to amino groups of proteins to give active conjugates. For example, the enzyme subtilisin (2 ml of a 2.37 mg/ml solution) was coupled to acryloyi-PEG-SPA (from Example 7) (MW 3400, 10 mg) by reaction in 2 ml of borate buffer (0.1 M, pH 8.0) for one hour at 4°C. The protein was purified by ultrafiltration with an Amicon PM 30 ultrafiltration membrane. Analysis with fluorescamine assay showed that two lysine groups had been modified by PEG attachment. Analysis on size exclusion chromatography (Toya Soda TSK 3000 column with pH 7 phosphate buffer eluent) showed that the molecular weight of the PEG-protein conjugate was approximately six thousand daltons greater than that of native protein. Exposure of the protein-PEG conjugate to a model substrate, the peptide PNA, gave rapid production of p-nitrophenol as shown by adsorbance at 402 nm, thus demonstrating that the protein-PEG conjugate has retained most of its catalytic activity.
In a similar experiment bovine alkaline phosphatase, MW 140,000, was coupled to methoxy-PEG- SPA, MW 5000, (from Example 1) by reaction of 20 mg of enzy e and 30 mg of the PEG in 2 ml of buffer containing 0.2 M sodium phosphate and 0.5 M NaCl (pH 7.5) at 4°C for 30 minutes. Unreacted PEG was removed by ultrafiltration, as above. Fluorescamine analysis showed that 20% of the available lysines were modified. Analysis on size exclusion chromatography (Toya Soda TSK 3000 column with pH 7 phosphate buffer eluent) showed that the molecular weight of the PEG-protein conjugate was approximately 30 thousand daltons greater than that of native protein. Addition of p- nitrophenylphosphate, a substrate for the enzyme, showed rapid production of p-nitrophenol (absorbance at 402 nm) , thus demonstrating that the protein-PEG conjugate has retained most of its catalytic activity. Other water soluble polymers than PEG should be suitable for similar modification to create the structures of the invention that are suitable for biotechnical use. These other polymers include poiy(vinyl alcohol) ("PVA") ; other poly(alkylene oxides) such as poly(propylene glycol) ("PPG") and the like; and poly(oxyethylated polyols) such as poly(oxyethylated glycerol) , poly(oxyethylated sorbitol) , and poly(oxyethylated glucose) , and the like. The polymers can be homopolymers or random or block copolymers and terpolymers based on the monomers of the above polymers, straight chain or branched, or substituted or unsubstituted similar to mPEG and other capped, monofunctional PEGs having a single active site available for attachment to a linker. Specific examples of suitable additional polymers include poly(oxazoline) , poly(acryloylmorpholine) ("PAcM") as described in published Italian Patent Application MI-92-A-0002616 filed November 17, 1992, and poly(vinylpyrrolidone) ("PVP") . PVP and poly(oxazoline) are well known polymers in the art and their preparation and use in the syntheses described above for mPEG should be readily apparent to the skilled artisan. The invention has been described with respect to several particular examples and embodiments. However, the foregoing examples and description are not intended to limit the invention to the exemplified embodiments, and the skilled artisan should recognize that variations can be made within the scope and spirit of the invention as described in the foregoing specification. The invention includes all alternatives, modifications, and equivalents that may be included within the true scope and spirit of the invention as defined by the appended claims.

Claims

WHAT IS CLAIMED IS: 1. An active ester of a polymer acid wherein said polymer acid has a single active ester and no other ester linkages, said active ester has a half life in water of from about 10 to 25 minutes, and said active ester comprises a propionic or butanoic acid moiety and a polymer moiety selected from the group consisting of poly(alkylene oxides) , poly(oxyethylated polyols) , poly(olefinic alcohols) , and poly(acrylomorpholine) .
2. The active ester of Claim 1 wherein said polymer moiety is poly(ethylene glycol) .
3. The active ester of Claim 2 wherein said poly(ethylene glycol) is represented by the general structure: -CH2CH20- (CH2CH20)n-CH2CH2- wherein n is from about 20 to 4000.
4. The active ester of Claim 3 wherein said active ester has the structure :
R- (OCH2CH2)n-Z- (CH2)m-C02-Q wherein n is from about 20 to 4000; Z is a hydrolytically stable moiety selected from the group consisting of -0-, -S-, -NHCO-, -CONH-, -NHC02-, and 02CNH- ; m is from 2 to 3 ; Q is selected from the group consisting of hydrogen, tert-butyl, N-succinimide, N- sulfosuccinimide, N-phthalimide, N-glutarimide, N- tetrahydrophthalimide, and N-norbornene-2, 3- dicarboximide, hydroxybenzotriazole, and hydroxy-7- azabenzotriazole; and R is selected from the group consisting of hydrogen, alkyl, benzyl, aryl, acetal (CH3CH20)2-CH2CH2-0-, aldehyde OHC-CH2CH2-0- , alkenyl CH2=CH-CH2-, acrylate CH2=CH-C0-, methacrylate CH2=C(CH3) -CO-, acrylamide CH2=CH-CONH-CH2CH2-, active sulfone, amine H2N-CH2CH2-, protected amine Y-NH-CH2CH2- , where Y is an amine protecting group, thiol HS-CH2-CH2, and protected thiol B-S-CH2CH2-, wherein B is a thiol protecting group.
5. The active ester of Claim 4 wherein n is from about 20 to 2000.
6. The active ester of Claim 4 wherein said amine protecting group is t-Boc and said thiol protecting group is orthopyridyldisulfide.
7. A compound having the following structure:
R-poly-Z- (CH2)m-C02-Q where poly represents a polymer moiety selected from the group consisting of poly(alkylene oxides), poly(oxyethylated polyols) , poly(olefinic alcohols) , and poly(acrylomorpholine) ; Z is a hydrolytically stable group having no ester linkages; m is from 2 to 3; Q is selected from the group consisting of hydrogen, tert-butyl, N-succinimide, N- sulfosuccinimide, N-phthalimide, N-glutarimide, N- tetrahydrophthalimide, and N-norbornene-2, 3- dicarboximide, hydroxybenzotriazole, and hydroxy-7- azabenzotriazole; and R is selected from the group consisting of hydrogen, alkyl, benzyl, aryl, acetal (CH3CH20)2-CH2CH2-0-, aldehyde OHC-CH2CH2-0- , alkenyl CH2=CH-CH2-, acrylate CH2=CH-C0-, methacrylate
CH2=C(CH3) -CO-, acrylamide CH2=CH-CONH-CH2CH2- , active sulfone, amine H2N-CH2CH2-, protected amine Y-NH-CH2CH2- , where Y is an amine protecting group, thiol HS-CH2-CH2, and protected thiol B-S-CH2CH2-, wherein B is a thiol protecting group.
8. A compound according to Claim 7 wherein Z is selected from the group consisting of -0-, -S-, -NHCO-, -CONH-, -NHC02-, and -02CNH- .
9. A compound according to Claim 7 wherein the amine protecting group Y is t-BOC and said thiol protecting group B is orthopyridyldisulide.
10. A compound according to Claim 7 wherein poly represents a polymer selected from the group consisting of poly(ethylene glycol) , poly(propylene glycol) , poly(oxyethylated glycerol) , poly(oxyethylated sorbitol), poly(oxyethylated glucose) , poly(vinyl alcohol), and poly(acrylomorpholine) .
11. A compound according to Claim 7 wherein the polymer backbone is substantially straight chain and unsubstituted except for the terminae.
12. A compound according to Claim 7 wherein the polymer backbone is a random or block copolymer or terpolymer.
13. A compound having the structure R- (OCH2CH2)n-0-CH2CH2-C02-NHS wherein n is from about 20 to 4000 and R is selected from the group consisting of hydrogen, alkyl, benzyl, aryl, acetal (CH3CH20) 2-CH2CH2-0- , aldehyde OHC-CH2CH2-0-, alkenyl CH2=CH-CH2-, acrylate CH2=CH-C0-, methacrylate CH2=C(CH3) -CO-, acrylamide CH2=CH-CONH-CH2CH2-, active sulfone, amine H2N-CH2CH2-, protected amine Y-NH-CH2CH2-, where Y is an amine protecting group, thiol HS-CH2-CH2, and protected thiol B-S-CH2CH2-, wherein B is a thiol protecting group.
14. The compound of Claim 13 wherein n is from about 20 to 2000.
15. A compound having the structure CH30-PEG-0-CH2CH2-C02-NHS.
16. A compound having the structure CH30-PEG-0-CH2CH2-CONH-PRO, wherein the moiety -NH-PRO represents an amino site on a biologically active molecule.
17. A water soluble, hydrolytically stable, and isolatable conjugate of a heterofunctional or monofunctional compound as claimed in Claim 1 and at least one biologically active molecule or a surface.
18. A conjugate according to Claim 17 wherein the at least one biologically active molecule is selected from the group consisting of proteins, enzymes, polypeptides, drugs, dyes, nucleosides, oligonucleotides, lipids, phospholipids, and liposomes
19. A conjugate according to Claim 17 wherein the compound serves to tether a biologically active molecule to another biologically active molecule, which may be the same or different, or to tether a biologically active molecule to a surface.
PCT/US1996/011261 1995-07-07 1996-07-03 Poly(ethylene glycol) and related polymers monosubstituted with propionic or butanoic acids and functional derivatives thereof for biotechnical applications WO1997003106A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU63457/96A AU6345796A (en) 1995-07-07 1996-07-03 Poly(ethylene glycol) and related polymers monosubstituted with propionic or butanoic acids and functional derivatives thereof for biotechnical applications

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US49932195A 1995-07-07 1995-07-07
US08/499,321 1995-07-07
US08/642,231 US5672662A (en) 1995-07-07 1995-10-02 Poly(ethylene glycol) and related polymers monosubstituted with propionic or butanoic acids and functional derivatives thereof for biotechnical applications
US08/642,231 1995-10-02

Publications (1)

Publication Number Publication Date
WO1997003106A1 true WO1997003106A1 (en) 1997-01-30

Family

ID=27053119

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1996/011261 WO1997003106A1 (en) 1995-07-07 1996-07-03 Poly(ethylene glycol) and related polymers monosubstituted with propionic or butanoic acids and functional derivatives thereof for biotechnical applications

Country Status (3)

Country Link
US (1) US5672662A (en)
AU (1) AU6345796A (en)
WO (1) WO1997003106A1 (en)

Cited By (86)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1999045964A1 (en) * 1998-03-12 1999-09-16 Shearwater Polymers, Incorporated Poly(ethylene glycol) derivatives with proximal reactive groups
WO2001026692A1 (en) * 1999-10-08 2001-04-19 Shearwater Corporation Heterobifunctional poly(ethylene glycol) derivatives and methods for their preparation
WO2001046291A1 (en) * 1999-12-22 2001-06-28 Shearwater Corporation Sterically hindered derivatives of water soluble polymers
WO2002049673A2 (en) 2000-12-20 2002-06-27 F. Hoffmann-La Roche Ag Conjugates of erythropoietin (pep) with polyethylene glycol (peg)
US6448369B1 (en) 1997-11-06 2002-09-10 Shearwater Corporation Heterobifunctional poly(ethylene glycol) derivatives and methods for their preparation
WO2003005026A2 (en) * 2001-07-02 2003-01-16 Amersham Biosciences Uk Limited Chemical capture reagent
WO2004000366A1 (en) 2002-06-21 2003-12-31 Novo Nordisk Health Care Ag Pegylated factor vii glycoforms
WO2004012773A1 (en) 2002-07-24 2004-02-12 F. Hoffmann-La Roche Ag Polyalkylene glycol acid additives
WO2004022629A2 (en) * 2002-09-09 2004-03-18 Nektar Therapeutics Al, Corporation Method for preparing water-soluble polymer derivatives bearing a terminal carboxylic acid
EP1423093A2 (en) * 2001-04-23 2004-06-02 Wisconsin Alumni Research Foundation Bifunctional-modified hydrogels
WO2004061094A1 (en) 2002-12-30 2004-07-22 Gryphon Therapeutics, Inc. Water-soluble thioester and selenoester compounds and methods for making and using the same
WO2005037214A2 (en) 2003-10-14 2005-04-28 Intermune, Inc. Macrocyclic carboxylic acids and acylsulfonamides as inhibitors of hcv replication
WO2006009901A2 (en) 2004-06-18 2006-01-26 Ambrx, Inc. Novel antigen-binding polypeptides and their uses
US7128913B2 (en) 2000-12-20 2006-10-31 Hoffmann-La Roche Inc. Erythropoietin conjugates
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
WO2007022780A1 (en) * 2005-08-26 2007-03-01 Novo Nordisk A/S A method of modifying a macromolecular system
WO2008030558A2 (en) 2006-09-08 2008-03-13 Ambrx, Inc. Modified human plasma polypeptide or fc scaffolds and their uses
EP1982732A2 (en) 2000-02-11 2008-10-22 Maxygen Holdings Ltd. Factor VII or VIIA-like molecules
WO2009067636A2 (en) 2007-11-20 2009-05-28 Ambrx, Inc. Modified insulin polypeptides and their uses
EP2080771A2 (en) 2001-02-27 2009-07-22 Maxygen Aps New interferon beta-like molecules
WO2009093713A1 (en) * 2008-01-25 2009-07-30 Ebara Corporation Peg-modified hydroxyapatite, pharmaceutical preparation containing the same as base material, and method for production of the same
EP2093235A1 (en) 2006-02-08 2009-08-26 Alios Biopharma Inc. Hyperglycosylated variants of interferon alfacon-1
US7608663B2 (en) 2004-01-21 2009-10-27 Nektar Therapeutics Method of preparing propionic acid-terminated polymers
EP2133098A1 (en) 2000-01-10 2009-12-16 Maxygen Holdings Ltd G-CSF conjugates
US7642323B2 (en) 1997-11-06 2010-01-05 Nektar Therapeutics Heterobifunctional poly(ethylene glycol) derivatives and methods for their preparation
WO2010007626A1 (en) * 2008-07-14 2010-01-21 Biocon Limited A method of synthesizing a substantially monodispersed mixture of oligomers
WO2010011735A2 (en) 2008-07-23 2010-01-28 Ambrx, Inc. Modified bovine g-csf polypeptides and their uses
US7736872B2 (en) 2004-12-22 2010-06-15 Ambrx, Inc. Compositions of aminoacyl-TRNA synthetase and uses thereof
EP2213733A2 (en) 2006-05-24 2010-08-04 Novo Nordisk Health Care AG Factor IX analogues having prolonged in vivo half life
US7816320B2 (en) 2004-12-22 2010-10-19 Ambrx, Inc. Formulations of human growth hormone comprising a non-naturally encoded amino acid at position 35
EP2263684A1 (en) 2003-10-10 2010-12-22 Novo Nordisk A/S IL-21 derivatives
EP2279756A2 (en) 2005-04-05 2011-02-02 Instituto di Ricerche di Biologia Molecolare p Angeletti S.P.A. Method for shielding functional sites or epitopes on proteins
EP2284191A2 (en) 2004-12-22 2011-02-16 Ambrx, Inc. Process for the preparation of hGH
US7947473B2 (en) 2004-12-22 2011-05-24 Ambrx, Inc. Methods for expression and purification of pegylated recombinant human growth hormone containing a non-naturally encoded keto amino acid
EP2327724A2 (en) 2004-02-02 2011-06-01 Ambrx, Inc. Modified human growth hormone polypeptides and their uses
US8012931B2 (en) 2007-03-30 2011-09-06 Ambrx, Inc. Modified FGF-21 polypeptides and their uses
WO2011107591A1 (en) 2010-03-05 2011-09-09 Rigshospitalet Chimeric inhibitor molecules of complement activation
WO2011143274A1 (en) 2010-05-10 2011-11-17 Perseid Therapeutics Polypeptide inhibitors of vla4
EP2390262A1 (en) 2003-05-16 2011-11-30 Intermune, Inc. Synthetic chemokine receptor ligands and methods of use thereof
US8093356B2 (en) 2005-06-03 2012-01-10 Ambrx, Inc. Pegylated human interferon polypeptides
US8114630B2 (en) 2007-05-02 2012-02-14 Ambrx, Inc. Modified interferon beta polypeptides and their uses
WO2012024452A2 (en) 2010-08-17 2012-02-23 Ambrx, Inc. Modified relaxin polypeptides and their uses
US8278418B2 (en) 2008-09-26 2012-10-02 Ambrx, Inc. Modified animal erythropoietin polypeptides and their uses
WO2013004607A1 (en) 2011-07-01 2013-01-10 Bayer Intellectual Property Gmbh Relaxin fusion polypeptides and uses thereof
WO2013006706A1 (en) 2011-07-05 2013-01-10 Bioasis Technologies Inc. P97-antibody conjugates and methods of use
EP2548967A2 (en) 2006-09-21 2013-01-23 The Regents of The University of California Aldehyde tags, uses thereof in site-specific protein modification
US8420792B2 (en) 2006-09-08 2013-04-16 Ambrx, Inc. Suppressor tRNA transcription in vertebrate cells
EP2633866A2 (en) 2003-10-17 2013-09-04 Novo Nordisk A/S Combination therapy
WO2013185115A1 (en) 2012-06-08 2013-12-12 Sutro Biopharma, Inc. Antibodies comprising site-specific non-natural amino acid residues, methods of their preparation and methods of their use
WO2013189745A2 (en) * 2012-06-18 2013-12-27 Basf Se Agroformulations containing a lactone based alkoxylate
WO2014022515A1 (en) 2012-07-31 2014-02-06 Bioasis Technologies, Inc. Dephosphorylated lysosomal storage disease proteins and methods of use thereof
WO2014036492A1 (en) 2012-08-31 2014-03-06 Sutro Biopharma, Inc. Modified amino acids comprising an azido group
WO2014160438A1 (en) 2013-03-13 2014-10-02 Bioasis Technologies Inc. Fragments of p97 and uses thereof
EP2805964A1 (en) 2009-12-21 2014-11-26 Ambrx, Inc. Modified bovine somatotropin polypeptides and their uses
EP2805965A1 (en) 2009-12-21 2014-11-26 Ambrx, Inc. Modified porcine somatotropin polypeptides and their uses
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
WO2015031673A2 (en) 2013-08-28 2015-03-05 Bioasis Technologies Inc. Cns-targeted conjugates having modified fc regions and methods of use thereof
WO2015054658A1 (en) 2013-10-11 2015-04-16 Sutro Biopharma, Inc. Modified amino acids comprising tetrazine functional groups, methods of preparation, and methods of their use
WO2015081282A1 (en) 2013-11-27 2015-06-04 Redwood Bioscience, Inc. Hydrazinyl-pyrrolo compounds and methods for producing a conjugate
US9121024B2 (en) 2008-09-26 2015-09-01 Ambrx, Inc. Non-natural amino acid replication-dependent microorganisms and vaccines
US9133495B2 (en) 2006-09-08 2015-09-15 Ambrx, Inc. Hybrid suppressor tRNA for vertebrate cells
US9310374B2 (en) 2012-11-16 2016-04-12 Redwood Bioscience, Inc. Hydrazinyl-indole compounds and methods for producing a conjugate
US9434778B2 (en) 2014-10-24 2016-09-06 Bristol-Myers Squibb Company Modified FGF-21 polypeptides comprising an internal deletion and uses thereof
US9488660B2 (en) 2005-11-16 2016-11-08 Ambrx, Inc. Methods and compositions comprising non-natural amino acids
EP3103880A1 (en) 2008-02-08 2016-12-14 Ambrx, Inc. Modified leptin polypeptides and their uses
US9567386B2 (en) 2010-08-17 2017-02-14 Ambrx, Inc. Therapeutic uses of modified relaxin polypeptides
US9579390B2 (en) 2012-11-12 2017-02-28 Redwood Bioscience, Inc. Compounds and methods for producing a conjugate
EP3135690A1 (en) 2012-06-26 2017-03-01 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
US9605078B2 (en) 2012-11-16 2017-03-28 The Regents Of The University Of California Pictet-Spengler ligation for protein chemical modification
US9920106B2 (en) 2003-12-18 2018-03-20 Novo Nordisk A/S GLP-1 compounds
US10266578B2 (en) 2017-02-08 2019-04-23 Bristol-Myers Squibb Company Modified relaxin polypeptides comprising a pharmacokinetic enhancer and uses thereof
WO2019133399A1 (en) 2017-12-26 2019-07-04 Becton, Dickinson And Company Deep ultraviolet-excitable water-solvated polymeric dyes
WO2019191482A1 (en) 2018-03-30 2019-10-03 Becton, Dickinson And Company Water-soluble polymeric dyes having pendant chromophores
WO2020023300A1 (en) 2018-07-22 2020-01-30 Bioasis Technologies, Inc. Treatment of lymmphatic metastases
WO2020056066A1 (en) 2018-09-11 2020-03-19 Ambrx, Inc. Interleukin-2 polypeptide conjugates and their uses
WO2020082057A1 (en) 2018-10-19 2020-04-23 Ambrx, Inc. Interleukin-10 polypeptide conjugates, dimers thereof, and their uses
WO2020168017A1 (en) 2019-02-12 2020-08-20 Ambrx, Inc. Compositions containing, methods and uses of antibody-tlr agonist conjugates
WO2021183832A1 (en) 2020-03-11 2021-09-16 Ambrx, Inc. Interleukin-2 polypeptide conjugates and methods of use thereof
WO2021236526A1 (en) 2020-05-18 2021-11-25 Bioasis Technologies, Inc. Compositions and methods for treating lewy body dementia
WO2021255524A1 (en) 2020-06-17 2021-12-23 Bioasis Technologies, Inc. Compositions and methods for treating frontotemporal dementia
WO2022040596A1 (en) 2020-08-20 2022-02-24 Ambrx, Inc. Antibody-tlr agonist conjugates, methods and uses thereof
US11273202B2 (en) 2010-09-23 2022-03-15 Elanco Us Inc. Formulations for bovine granulocyte colony stimulating factor and variants thereof
WO2022212899A1 (en) 2021-04-03 2022-10-06 Ambrx, Inc. Anti-her2 antibody-drug conjugates and uses thereof
EP4155349A1 (en) 2021-09-24 2023-03-29 Becton, Dickinson and Company Water-soluble yellow green absorbing dyes
WO2024007016A2 (en) 2022-07-01 2024-01-04 Beckman Coulter, Inc. Novel fluorescent dyes and polymers from dihydrophenanthrene derivatives
WO2024044327A1 (en) 2022-08-26 2024-02-29 Beckman Coulter, Inc. Dhnt monomers and polymer dyes with modified photophysical properties

Families Citing this family (417)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20020177688A1 (en) * 1988-12-22 2002-11-28 Kirin-Amgen, Inc., Chemically-modified G-CSF
EP0401384B1 (en) * 1988-12-22 1996-03-13 Kirin-Amgen, Inc. Chemically modified granulocyte colony stimulating factor
KR100361933B1 (en) 1993-09-08 2003-02-14 라 졸라 파마슈티칼 컴파니 Chemically defined nonpolymeric bonds form the platform molecule and its conjugate
US6057287A (en) 1994-01-11 2000-05-02 Dyax Corp. Kallikrein-binding "Kunitz domain" proteins and analogues thereof
US20030053982A1 (en) * 1994-09-26 2003-03-20 Kinstler Olaf B. N-terminally chemically modified protein compositions and methods
US5824784A (en) * 1994-10-12 1998-10-20 Amgen Inc. N-terminally chemically modified protein compositions and methods
US5795587A (en) 1995-01-23 1998-08-18 University Of Pittsburgh Stable lipid-comprising drug delivery complexes and methods for their production
US6008202A (en) * 1995-01-23 1999-12-28 University Of Pittsburgh Stable lipid-comprising drug delivery complexes and methods for their production
US20040266706A1 (en) * 2002-11-05 2004-12-30 Muthiah Manoharan Cross-linked oligomeric compounds and their use in gene modulation
US5898031A (en) 1996-06-06 1999-04-27 Isis Pharmaceuticals, Inc. Oligoribonucleotides for cleaving RNA
US9096636B2 (en) 1996-06-06 2015-08-04 Isis Pharmaceuticals, Inc. Chimeric oligomeric compounds and their use in gene modulation
US7812149B2 (en) 1996-06-06 2010-10-12 Isis Pharmaceuticals, Inc. 2′-Fluoro substituted oligomeric compounds and compositions for use in gene modulations
WO2005121371A2 (en) * 2004-06-03 2005-12-22 Isis Pharmaceuticals, Inc. Double strand compositions comprising differentially modified strands for use in gene modulation
US7091311B2 (en) * 1996-06-07 2006-08-15 Smithkline Beecham Corporation Peptides and compounds that bind to a receptor
US6214966B1 (en) * 1996-09-26 2001-04-10 Shearwater Corporation Soluble, degradable poly(ethylene glycol) derivatives for controllable release of bound molecules into solution
US6743248B2 (en) 1996-12-18 2004-06-01 Neomend, Inc. Pretreatment method for enhancing tissue adhesion
US20040176801A1 (en) * 1997-03-12 2004-09-09 Neomend, Inc. Pretreatment method for enhancing tissue adhesion
US20030191496A1 (en) * 1997-03-12 2003-10-09 Neomend, Inc. Vascular sealing device with microwave antenna
US6371975B2 (en) 1998-11-06 2002-04-16 Neomend, Inc. Compositions, systems, and methods for creating in situ, chemically cross-linked, mechanical barriers
US6168784B1 (en) 1997-09-03 2001-01-02 Gryphon Sciences N-terminal modifications of RANTES and methods of use
US20050158273A1 (en) * 1997-09-25 2005-07-21 Harris J. M. Soluble, degradable polyethylene glycol) derivatives for controllable release of bound molecules into solution
US7347850B2 (en) * 1998-08-14 2008-03-25 Incept Llc Adhesion barriers applicable by minimally invasive surgery and methods of use thereof
US6458147B1 (en) 1998-11-06 2002-10-01 Neomend, Inc. Compositions, systems, and methods for arresting or controlling bleeding or fluid leakage in body tissue
US6994686B2 (en) 1998-08-26 2006-02-07 Neomend, Inc. Systems for applying cross-linked mechanical barriers
DK1400551T3 (en) 1998-08-28 2007-10-15 Amylin Pharmaceuticals Inc Precise length polyamide chains and their proteins conjugates
IL142350A0 (en) * 1998-10-16 2002-03-10 Biogen Inc Interferon-beta fusion proteins and pharmaceutical compositions containing the same
SG2008070138A (en) * 1998-10-16 2017-08-30 Biogen Ma Inc Polymer conjugates of interferon beta- 1a and their uses
US6830756B2 (en) * 1998-11-06 2004-12-14 Neomend, Inc. Systems, methods, and compositions for achieving closure of vascular puncture sites
US6949114B2 (en) 1998-11-06 2005-09-27 Neomend, Inc. Systems, methods, and compositions for achieving closure of vascular puncture sites
US7279001B2 (en) * 1998-11-06 2007-10-09 Neomend, Inc. Systems, methods, and compositions for achieving closure of vascular puncture sites
US6899889B1 (en) * 1998-11-06 2005-05-31 Neomend, Inc. Biocompatible material composition adaptable to diverse therapeutic indications
US20080114092A1 (en) * 1998-12-04 2008-05-15 Incept Llc Adhesion barriers applicable by minimally invasive surgery and methods of use thereof
US6458953B1 (en) * 1998-12-09 2002-10-01 La Jolla Pharmaceutical Company Valency platform molecules comprising carbamate linkages
ATE246202T1 (en) * 1999-01-29 2003-08-15 Hoffmann La Roche GCSF CONJUGATES
US6270806B1 (en) 1999-03-03 2001-08-07 Elan Pharma International Limited Use of peg-derivatized lipids as surface stabilizers for nanoparticulate compositions
AU779887B2 (en) * 1999-06-08 2005-02-17 La Jolla Pharmaceutical Company Valency platform molecules comprising aminooxy groups
CZ299516B6 (en) 1999-07-02 2008-08-20 F. Hoffmann-La Roche Ag Erythropoietin glycoprotein conjugate, process for its preparation and use and pharmaceutical composition containing thereof
US7008635B1 (en) 1999-09-10 2006-03-07 Genzyme Corporation Hydrogels for orthopedic repair
US6348558B1 (en) 1999-12-10 2002-02-19 Shearwater Corporation Hydrolytically degradable polymers and hydrogels made therefrom
US7074878B1 (en) * 1999-12-10 2006-07-11 Harris J Milton Hydrolytically degradable polymers and hydrogels made therefrom
WO2001093914A2 (en) 2000-06-08 2001-12-13 La Jolla Pharmaceutical Company Multivalent platform molecules comprising high molecular weight polyethylene oxide
CN1460023A (en) * 2000-07-12 2003-12-03 格莱风治疗公司 Polymer-modified bioactive sythetic chemokines, and method for their mfg. and use
US7118737B2 (en) * 2000-09-08 2006-10-10 Amylin Pharmaceuticals, Inc. Polymer-modified synthetic proteins
AU7338801A (en) * 2000-09-08 2002-03-22 Gryphon Sciences "pseudo"-native chemical ligation
US20020065397A1 (en) 2000-10-12 2002-05-30 Joseph Roberts Protecting therapeutic compositions from host-mediated inactivation
JP2004534721A (en) * 2000-10-31 2004-11-18 ピーアール ファーマシューティカルズ,インク. Methods and compositions for enhanced delivery of bioactive molecules
US7053150B2 (en) * 2000-12-18 2006-05-30 Nektar Therapeutics Al, Corporation Segmented polymers and their conjugates
TW593427B (en) * 2000-12-18 2004-06-21 Nektar Therapeutics Al Corp Synthesis of high molecular weight non-peptidic polymer derivatives
US9700866B2 (en) * 2000-12-22 2017-07-11 Baxter International Inc. Surfactant systems for delivery of organic compounds
US7265186B2 (en) * 2001-01-19 2007-09-04 Nektar Therapeutics Al, Corporation Multi-arm block copolymers as drug delivery vehicles
TWI246524B (en) * 2001-01-19 2006-01-01 Shearwater Corp Multi-arm block copolymers as drug delivery vehicles
EP1370249A4 (en) * 2001-02-26 2006-05-03 Univ Duke Novel dendriticpolymers and their biomedical uses
US20050276858A1 (en) * 2001-04-23 2005-12-15 Kao Weiyuan J Bifunctional-modified hydrogels
AU2002256398A2 (en) * 2001-04-30 2002-11-11 Targeted Genetics Corporation Lipid-comprising drug delivery complexes and methods for their production
EP1421175B1 (en) * 2001-06-28 2008-11-19 Mountain View Pharmaceuticals, Inc. Polymer stabilized proteinases
US7009033B2 (en) * 2001-07-02 2006-03-07 Polymer Source Inc. Heterofunctional polyethylene glycol and polyethylene oxide, process for their manufacture
US20040077835A1 (en) * 2001-07-12 2004-04-22 Robin Offord Chemokine receptor modulators, production and use
US20040208844A1 (en) * 2001-08-01 2004-10-21 Francis Ignatious Products and drug delivery vehicles
US7214660B2 (en) 2001-10-10 2007-05-08 Neose Technologies, Inc. Erythropoietin: remodeling and glycoconjugation of erythropoietin
WO2004099231A2 (en) 2003-04-09 2004-11-18 Neose Technologies, Inc. Glycopegylation methods and proteins/peptides produced by the methods
EP2322229B1 (en) 2001-10-10 2016-12-21 Novo Nordisk A/S Remodeling and glycoconjugation of Factor IX
US7173003B2 (en) 2001-10-10 2007-02-06 Neose Technologies, Inc. Granulocyte colony stimulating factor: remodeling and glycoconjugation of G-CSF
US7157277B2 (en) 2001-11-28 2007-01-02 Neose Technologies, Inc. Factor VIII remodeling and glycoconjugation of Factor VIII
EP2279755B1 (en) 2001-10-10 2014-02-26 ratiopharm GmbH Remodelling and glycoconjugation of Fibroblast Growth Factor (FGF)
US8008252B2 (en) 2001-10-10 2011-08-30 Novo Nordisk A/S Factor VII: remodeling and glycoconjugation of Factor VII
PT1436012T (en) 2001-10-18 2018-03-27 Nektar Therapeutics Polymer conjugates of opioid antagonists
AU2002346686A1 (en) * 2001-12-07 2003-06-23 Intermune, Inc. Compositions and method for treating hepatitis virus infection
ES2566797T3 (en) * 2002-01-18 2016-04-15 Biogen Ma Inc. Polyalkylene polymer compounds and uses thereof
US20040131582A1 (en) * 2002-02-26 2004-07-08 Grinstaff Mark W. Novel dendritic polymers and their biomedical uses
CN100475269C (en) * 2002-03-05 2009-04-08 北京键凯科技有限公司 Binding agent of hydrophilic polymer-glutamic acid oligopeptide and medicinal molecular, composition containing said binding agent and use thereof
DE60315145T2 (en) * 2002-03-13 2008-04-30 Beijing Jiankai Technology Co., Ltd. HYDROPHILIC Y-BRANCH POLYMER DERIVATIVE AND METHOD OF MANUFACTURING THEREOF; ABOVE CONNECTED MEDICAL COMPOSITE
US7153829B2 (en) 2002-06-07 2006-12-26 Dyax Corp. Kallikrein-inhibitor therapies
JP2005534647A (en) 2002-06-07 2005-11-17 ダイアックス、コープ Prevention and reduction of blood loss
MXPA04012496A (en) 2002-06-21 2005-09-12 Novo Nordisk Healthcare Ag Pegylated factor vii glycoforms.
RU2294938C2 (en) * 2002-07-24 2007-03-10 Ф.Хоффманн-Ля Рош Аг Peg-containing polypeptide t1249
CN100357317C (en) * 2002-07-24 2007-12-26 弗·哈夫曼-拉罗切有限公司 Pegylated T20 polypeptide
PL214871B1 (en) * 2002-07-24 2013-09-30 Hoffmann La Roche Polyethylene glycol aldehyde derivatives
US7459435B2 (en) * 2002-08-29 2008-12-02 Hoffmann-La Roche Inc. Treatment of disturbances of iron distribution
WO2004019991A2 (en) * 2002-08-30 2004-03-11 F. Hoffmann-La Roche Ag Scatter factor/hepatocyte growth factor antagonist nk4 for the treatment of glioma
AP2005003246A0 (en) 2002-09-09 2005-03-31 Nektar Therapeutics Al Corp Water-soluble polymer alkanals
US8067367B2 (en) 2002-09-18 2011-11-29 Janssen Pharmaceutica, N.V. Methods of increasing platelet and hematopoietic stem cell production
CA2498062C (en) * 2002-09-27 2010-05-25 F. Hoffmann-La Roche Ag Conjugates of insulin-like growth factor binding protein-4 and poly(ethylene glycol)
US20040062748A1 (en) 2002-09-30 2004-04-01 Mountain View Pharmaceuticals, Inc. Polymer conjugates with decreased antigenicity, methods of preparation and uses thereof
US8129330B2 (en) * 2002-09-30 2012-03-06 Mountain View Pharmaceuticals, Inc. Polymer conjugates with decreased antigenicity, methods of preparation and uses thereof
WO2004044139A2 (en) 2002-11-05 2004-05-27 Isis Parmaceuticals, Inc. Modified oligonucleotides for use in rna interference
TWI281864B (en) * 2002-11-20 2007-06-01 Pharmacia Corp N-terminally monopegylated human growth hormone conjugates and process for their preparation
US7459436B2 (en) * 2002-11-22 2008-12-02 Hoffmann-La Roche Inc. Treatment of disturbances of iron distribution
MXPA05006944A (en) * 2002-12-26 2005-12-14 Mountain View Pharmaceuticals Polymer conjugates of interferon-beta.
RS20050501A (en) * 2002-12-26 2007-08-03 Mountain View Pharmaceuticals Inc., Polymer conjugates of cytokines,chemokines,growth factors, polypeptide hormones and antagonists thereof with preserved receptor-binding activity
WO2004060967A1 (en) * 2002-12-31 2004-07-22 Nektar Therapeutics Al, Corporation Methods for the formation of hydrogels using thiosulfonate compositions and uses thereof
AU2004204136B2 (en) 2003-01-06 2008-10-09 Nektar Therapeutics Thiol-selective water-soluble polmer derivatives
US20050221443A1 (en) * 2003-01-06 2005-10-06 Xencor, Inc. Tumor necrosis factor super family agonists
US20060014248A1 (en) * 2003-01-06 2006-01-19 Xencor, Inc. TNF super family members with altered immunogenicity
US20050130892A1 (en) * 2003-03-07 2005-06-16 Xencor, Inc. BAFF variants and methods thereof
US7553930B2 (en) * 2003-01-06 2009-06-30 Xencor, Inc. BAFF variants and methods thereof
US8663650B2 (en) 2003-02-21 2014-03-04 Ac Immune Sa Methods and compositions comprising supramolecular constructs
US20090123367A1 (en) * 2003-03-05 2009-05-14 Delfmems Soluble Glycosaminoglycanases and Methods of Preparing and Using Soluble Glycosaminoglycanases
DE602004024041D1 (en) * 2003-03-05 2009-12-24 Halozyme Inc SOLUBLE HYALURONIDASE GLYCOPROTEIN (SHASEGP), METHOD OF ITS PREPARATION, USES AND PHARMACEUTICAL COMPOSITIONS CONTAINING THEREOF
US7871607B2 (en) * 2003-03-05 2011-01-18 Halozyme, Inc. Soluble glycosaminoglycanases and methods of preparing and using soluble glycosaminoglycanases
US20060104968A1 (en) * 2003-03-05 2006-05-18 Halozyme, Inc. Soluble glycosaminoglycanases and methods of preparing and using soluble glycosaminogly ycanases
JP2006523211A (en) 2003-03-14 2006-10-12 ネオス テクノロジーズ インコーポレイテッド Branched water-soluble polymers and their composites
US7642340B2 (en) 2003-03-31 2010-01-05 Xencor, Inc. PEGylated TNF-α variant proteins
US7610156B2 (en) * 2003-03-31 2009-10-27 Xencor, Inc. Methods for rational pegylation of proteins
CA2520875A1 (en) * 2003-03-31 2004-10-21 Xencor, Inc. Methods for rational pegylation of proteins
US8791070B2 (en) 2003-04-09 2014-07-29 Novo Nordisk A/S Glycopegylated factor IX
AU2004229461A1 (en) 2003-04-11 2004-10-28 Pr Pharmaceuticals Inc. Method for preparation of site-specific protein conjugates
WO2004091517A2 (en) 2003-04-15 2004-10-28 Smithkline Beecham Corporation Conjugates comprising human il-18 and substitution mutants thereof
EP1624847B1 (en) 2003-05-09 2012-01-04 BioGeneriX AG Compositions and methods for the preparation of human growth hormone glycosylation mutants
NZ563042A (en) * 2003-05-12 2008-09-26 Affymax Inc Novel spacer moiety for poly(ethylene glycol)-modified peptide-based compounds
PL1625156T3 (en) * 2003-05-12 2013-03-29 Affymax Inc Peptides that bind to the erythropoietin receptor
CN1820024B (en) 2003-05-12 2011-06-22 阿费麦克斯公司 Novel poly(ethylene glycol) modified compounds and uses thereof
US7459146B2 (en) 2003-05-30 2008-12-02 3M Innovative Properties Company Stabilized aerosol dispersions
US7109247B2 (en) * 2003-05-30 2006-09-19 3M Innovative Properties Company Stabilized particle dispersions containing nanoparticles
US20040249119A1 (en) * 2003-06-05 2004-12-09 Fox Martin Edward Novel mPEG propionaldehyde precursor
US8604159B2 (en) 2003-07-22 2013-12-10 Nektar Therapeutics Method for preparing functionalized polymers from polymer alcohols
US9005625B2 (en) 2003-07-25 2015-04-14 Novo Nordisk A/S Antibody toxin conjugates
US20050255561A1 (en) * 2003-07-25 2005-11-17 Pierre-Francois Tosi Therapeutic vaccine targeted against P-glycoprotein 170 for inhibiting multidrug resistance in the treatment of cancers
WO2005021557A2 (en) * 2003-08-29 2005-03-10 Dyax Corp. Poly-pegylated protease inhibitors
CA2537336C (en) 2003-09-17 2013-02-26 Nektar Therapeutics Al, Corporation Multi-arm polymer prodrugs
EP1675620B1 (en) * 2003-10-09 2019-05-08 Ambrx, Inc. Polymer derivatives
WO2005035564A2 (en) 2003-10-10 2005-04-21 Xencor, Inc. Protein based tnf-alpha variants for the treatment of tnf-alpha related disorders
WO2005042563A2 (en) * 2003-10-22 2005-05-12 Akzo Nobel N.V. Process for incrasing protein pegylation reaction yields by diafiltration ultrafiltration
US7407973B2 (en) * 2003-10-24 2008-08-05 Intermune, Inc. Use of pirfenidone in therapeutic regimens
ATE524203T1 (en) * 2003-11-03 2011-09-15 Medtronic Inc HYDROGEL FOR CELL-SPECIFIC INGROWTH
WO2005044836A2 (en) 2003-11-05 2005-05-19 Genovoxx Gmbh Macromolecular nucleotide compounds and methods for using the same
US20050214250A1 (en) 2003-11-06 2005-09-29 Harris J M Method of preparing carboxylic acid functionalized polymers
US8633157B2 (en) 2003-11-24 2014-01-21 Novo Nordisk A/S Glycopegylated erythropoietin
US20080305992A1 (en) 2003-11-24 2008-12-11 Neose Technologies, Inc. Glycopegylated erythropoietin
US7189768B2 (en) * 2003-11-25 2007-03-13 3M Innovative Properties Company Solution containing surface-modified nanoparticles
US7956032B2 (en) 2003-12-03 2011-06-07 Novo Nordisk A/S Glycopegylated granulocyte colony stimulating factor
US20060040856A1 (en) * 2003-12-03 2006-02-23 Neose Technologies, Inc. Glycopegylated factor IX
EP2905033B1 (en) 2003-12-16 2020-09-02 Nektar Therapeutics Monodisperse PEGylated naloxol compositions
PL1696947T3 (en) * 2003-12-19 2014-08-29 Hoffmann La Roche Use of erythropoietin in the treatment of disturbances of iron distribution in chronic inflammatory intestinal diseases
ES2560657T3 (en) * 2004-01-08 2016-02-22 Ratiopharm Gmbh O-linked glycosylation of G-CSF peptides
US6887952B1 (en) * 2004-02-12 2005-05-03 Biosite, Inc. N-aryl-carbamic acid ester-derived and valeric acid ester-derived cross-linkers and conjugates, and methods for their synthesis and use
US8569474B2 (en) 2004-03-09 2013-10-29 Isis Pharmaceuticals, Inc. Double stranded constructs comprising one or more short strands hybridized to a longer strand
WO2005115477A2 (en) * 2004-04-13 2005-12-08 Quintessence Biosciences, Inc. Non-natural ribonuclease conjugates as cytotoxic agents
US8562965B2 (en) 2004-05-03 2013-10-22 Nektar Therapeutics Polymer derivatives comprising an acetal or ketal branching point
US8394947B2 (en) 2004-06-03 2013-03-12 Isis Pharmaceuticals, Inc. Positionally modified siRNA constructs
US20060018875A1 (en) * 2004-06-14 2006-01-26 Blatt Lawrence M Interferon compositions and methods of use thereof
US20080300173A1 (en) 2004-07-13 2008-12-04 Defrees Shawn Branched Peg Remodeling and Glycosylation of Glucagon-Like Peptides-1 [Glp-1]
AU2005327906B2 (en) 2004-07-21 2010-05-13 Ambrx, Inc. Biosynthetic polypeptides utilizing non-naturally encoded amino acids
US20060040377A1 (en) * 2004-08-17 2006-02-23 Biocept, Inc. Protein microarrays
US7884086B2 (en) 2004-09-08 2011-02-08 Isis Pharmaceuticals, Inc. Conjugates for use in hepatocyte free uptake assays
EP1794588A2 (en) * 2004-09-09 2007-06-13 Biosite Incorporated Methods and compositions for measuring canine bnp and uses thereof
WO2006031811A2 (en) 2004-09-10 2006-03-23 Neose Technologies, Inc. Glycopegylated interferon alpha
US7235530B2 (en) 2004-09-27 2007-06-26 Dyax Corporation Kallikrein inhibitors and anti-thrombolytic agents and uses thereof
EP2586456B1 (en) 2004-10-29 2016-01-20 ratiopharm GmbH Remodeling and glycopegylation of fibroblast growth factor (FGF)
EP1814910A4 (en) * 2004-11-11 2009-04-29 Affymax Inc Novel peptides that bind to the erythropoietin receptor
WO2006062685A2 (en) * 2004-11-11 2006-06-15 Affymax, Inc. Novel peptides that bind to the erythropoietin receptor
EP1674113A1 (en) 2004-12-22 2006-06-28 F. Hoffmann-La Roche Ag Conjugates of insulin-like growth factor-1 (IGF-1) and poly(ethylene glycol)
GB2438982A (en) 2004-12-22 2007-12-12 Ambrx Inc Compostions containing, methods involving, and uses of non-natural amino acids and polypeptides
CA2593682C (en) 2005-01-10 2016-03-22 Neose Technologies, Inc. Glycopegylated granulocyte colony stimulating factor
WO2006078813A2 (en) * 2005-01-21 2006-07-27 Biosite Incorporated Arginine analogs, and methods for their synthesis and use
KR100645852B1 (en) 2005-01-22 2006-11-14 나재식 Hydrophillic quenching solution containing fatty acid grafted polyalkylene glycol
US20060233740A1 (en) * 2005-03-23 2006-10-19 Bossard Mary J Conjugates of an hGH moiety and a polymer
US20060222596A1 (en) 2005-04-01 2006-10-05 Trivascular, Inc. Non-degradable, low swelling, water soluble radiopaque hydrogel polymer
US20070154992A1 (en) * 2005-04-08 2007-07-05 Neose Technologies, Inc. Compositions and methods for the preparation of protease resistant human growth hormone glycosylation mutants
WO2006127910A2 (en) 2005-05-25 2006-11-30 Neose Technologies, Inc. Glycopegylated erythropoietin formulations
US20080255026A1 (en) * 2005-05-25 2008-10-16 Glycopegylated Factor 1X Glycopegylated Factor Ix
US7919461B2 (en) 2005-06-03 2011-04-05 Affymax, Inc. Erythropoietin receptor peptide formulations and uses
US8324159B2 (en) * 2005-06-03 2012-12-04 Affymax, Inc. Erythropoietin receptor peptide formulations and uses
US7550433B2 (en) * 2005-06-03 2009-06-23 Affymax, Inc. Erythropoietin receptor peptide formulations and uses
EP1926768B1 (en) * 2005-07-18 2011-09-14 Nektar Therapeutics Branched functionalized polymers using branched polyol cores
US7632823B2 (en) 2005-08-18 2009-12-15 Ambrx, Inc. Compositions of tRNA and uses thereof
US20070105755A1 (en) 2005-10-26 2007-05-10 Neose Technologies, Inc. One pot desialylation and glycopegylation of therapeutic peptides
DE102005041570A1 (en) 2005-09-01 2007-03-22 Celares Gmbh Highly branched reagents for modifaction of biopharmaceuticals, their preparation and use
US20070072795A1 (en) * 2005-09-28 2007-03-29 Anton Haselbeck Treatment of neurodegenerative disorders
WO2007047303A2 (en) * 2005-10-12 2007-04-26 Alvine Pharmaceuticals, Inc. Pegylated glutenase polypeptides
WO2007047922A2 (en) * 2005-10-19 2007-04-26 Smartcells, Inc. Polymer-drug conjugates
WO2007056191A2 (en) 2005-11-03 2007-05-18 Neose Technologies, Inc. Nucleotide sugar purification using membranes
CN101400646A (en) * 2005-11-08 2009-04-01 Ambrx公司 Accelerants for the modification of non-natural amino acids and non-natural amino acid polypeptides
NZ568578A (en) * 2005-12-14 2011-10-28 Ambrx Inc Compositions containing, methods involving, and uses of non-natural amino acids and polypeptides
CA2635948C (en) * 2006-01-16 2015-02-10 Bwe I Malmo Ab Low protonation constant hydroxy acids
US20070173634A1 (en) * 2006-01-24 2007-07-26 Olsson Nils U Methods for one-step purification of organic polymers using tangential flow filtration
JP5312050B2 (en) 2006-02-21 2013-10-09 ネクター セラピューティクス Split-type degradable polymers and composites produced therefrom
US8795709B2 (en) 2006-03-29 2014-08-05 Incept Llc Superabsorbent, freeze dried hydrogels for medical applications
EP2004633A4 (en) * 2006-03-30 2009-08-26 Palatin Technologies Inc Linear natriuretic peptide constructs
AU2007233116B2 (en) * 2006-03-30 2013-05-09 Palatin Technologies, Inc. Cyclic natriuretic peptide constructs
US8580746B2 (en) * 2006-03-30 2013-11-12 Palatin Technologies, Inc. Amide linkage cyclic natriuretic peptide constructs
WO2007117685A2 (en) 2006-04-07 2007-10-18 Nektar Therapeutics Al, Corporation Conjugates of an anti-tnf-alpha antibody
US8183376B2 (en) 2006-04-21 2012-05-22 Nektar Therapeutics Stereoselective reduction of a morphinone
US20100119697A1 (en) * 2006-05-10 2010-05-13 3M Innovative Properties Company Compositions and coatings containing fluorescent, inorganic nanoparticles
US7872068B2 (en) * 2006-05-30 2011-01-18 Incept Llc Materials formable in situ within a medical device
WO2008002482A2 (en) * 2006-06-23 2008-01-03 Surmodics, Inc. Hydrogel-based joint repair system and method
WO2007149594A2 (en) 2006-06-23 2007-12-27 Quintessence Biosciences, Inc. Modified ribonucleases
JP2009543868A (en) 2006-07-17 2009-12-10 クインテセンス バイオサイエンシーズ インコーポレーティッド Methods and compositions for cancer treatment
US20080280818A1 (en) 2006-07-21 2008-11-13 Neose Technologies, Inc. Glycosylation of peptides via o-linked glycosylation sequences
BRPI0715754A2 (en) * 2006-08-31 2013-07-09 Hoffmann La Roche Method for the production of insulin-like growth factor
CL2007002502A1 (en) 2006-08-31 2008-05-30 Hoffmann La Roche VARIANTS OF THE SIMILAR GROWTH FACTOR TO HUMAN INSULIN-1 (IGF-1) PEGILATED IN LISIN; METHOD OF PRODUCTION; FUSION PROTEIN THAT UNDERSTANDS IT; AND ITS USE TO TREAT ALZHEIMER'S DISEASE.
WO2008057683A2 (en) 2006-10-03 2008-05-15 Novo Nordisk A/S Methods for the purification of polypeptide conjugates
JP5457185B2 (en) 2006-10-04 2014-04-02 ノヴォ ノルディスク アー/エス Glycerol-linked PEGylated sugars and glycopeptides
JP2010520855A (en) * 2007-01-31 2010-06-17 アフィーマックス・インコーポレイテッド Nitrogen-based linkers for attaching modifying groups to polypeptides and other macromolecules
WO2008101311A1 (en) * 2007-02-22 2008-08-28 Biovectra Inc. Process for purification of water soluble polymers
US20140011964A1 (en) 2007-02-28 2014-01-09 Serina Therapeutics, Inc. Activated Polyoxazolines and Conjugates and Compositions Comprising the Same
EP2134181A4 (en) * 2007-02-28 2011-09-28 Serina Therapeutics Inc Activated polyoxazolines and compositions comprising the same
US8389759B2 (en) 2007-03-12 2013-03-05 Nektar Therapeutics Oligomer-anticholinergic agent conjugates
AU2008226820A1 (en) * 2007-03-12 2008-09-18 Nektar Therapeutics De novo synthesis of conjugates
EP2500039A1 (en) 2007-03-12 2012-09-19 Nektar Therapeutics Oligomer-antihistamine conjugates
US10512644B2 (en) 2007-03-12 2019-12-24 Inheris Pharmaceuticals, Inc. Oligomer-opioid agonist conjugates
PL2134371T3 (en) 2007-03-12 2015-06-30 Nektar Therapeutics Oligomer-opioid agonist conjugates
EP2131865B1 (en) 2007-03-12 2014-12-17 Nektar Therapeutics Oligomer-protease inhibitor conjugates
WO2008112287A1 (en) 2007-03-12 2008-09-18 Nektar Therapeutics Oligomer-beta blocker conjugates
US8173666B2 (en) 2007-03-12 2012-05-08 Nektar Therapeutics Oligomer-opioid agonist conjugates
BRPI0809670A8 (en) 2007-04-03 2018-12-18 Biogenerix Ag methods to increase stem cell production, to increase the number of granulocytes in an individual, to prevent, treat and alleviate myelosuppression that results from cancer therapy, to treat a condition in an individual, to treat neutropenia and thrombocytopenia in a mammal, to expand hematopoietic stem cells in culture, to increase hematopoiesis in an individual, to increase the number of hematopoietic progenitor cells in an individual, and to provide stable bone marrow graft, and, oral dosage form.
US20080287633A1 (en) * 2007-05-18 2008-11-20 Drumheller Paul D Hydrogel Materials
EP2150568B1 (en) * 2007-05-29 2016-03-23 Youl Chon Chemical Co. Ltd. Chain-end functionalized methoxy poly(ethylene glycol)and metal nano-particles using the same
MX2009013259A (en) 2007-06-12 2010-01-25 Novo Nordisk As Improved process for the production of nucleotide sugars.
CN101357986B (en) * 2007-06-13 2013-06-05 康达医药科技有限公司 Polyethylene glycol functional derivatives and manufacturing method thereof
WO2009002873A1 (en) * 2007-06-22 2008-12-31 Cvi Pharmaceuticals Limited Compounds, compositions and methods for reducing lipid levels
WO2009026334A2 (en) * 2007-08-21 2009-02-26 Genzyme Corporation Treatment with kallikrein inhibitors
US8207112B2 (en) 2007-08-29 2012-06-26 Biogenerix Ag Liquid formulation of G-CSF conjugate
US8748648B2 (en) * 2007-09-06 2014-06-10 Nektar Therapeutics Oligomer-calcium channel blocker conjugates
WO2009042064A2 (en) 2007-09-21 2009-04-02 Nektar Therapeutics Al, Corporation Oligomer-nucleoside phosphate conjugates
WO2009045539A2 (en) 2007-10-05 2009-04-09 Nektar Therapeutics Al, Corporation Oligomer-corticosteroid conjugates
WO2009045536A2 (en) * 2007-10-05 2009-04-09 The University Of North Carolina At Chapel Hill Receptor targeted oligonucleotides
CA2702043A1 (en) * 2007-10-08 2009-04-16 Quintessence Biosciences, Inc. Compositions and methods for ribonuclease-based therapies
US20100303753A1 (en) * 2007-10-19 2010-12-02 Nektar Therapeutics Oligomer Conjugates of Lidocaine and Its Derivatives
US8440787B2 (en) * 2007-10-23 2013-05-14 Nektar Therapeutics Hydroxyapatite-targeting multiarm polymers and conjugates made therefrom
US8853247B2 (en) * 2007-11-02 2014-10-07 Nektar Therapeutics Oligomer-nitroimidazole anti-infective conjugates
US8173597B2 (en) * 2007-11-09 2012-05-08 Baxter International Inc. Modified recombinant factor VIII and von Willebrand factor and methods of use
US20090123519A1 (en) * 2007-11-12 2009-05-14 Surmodics, Inc. Swellable hydrogel matrix and methods
US8536213B2 (en) * 2007-11-16 2013-09-17 Nektar Therapeutics Oligomer-dantrolene conjugates and related compounds
WO2009073154A1 (en) 2007-11-28 2009-06-11 Nektar Therapeutics Al, Corporation Oligomer-tricyclic conjugates
WO2009089053A1 (en) * 2008-01-11 2009-07-16 Nektar Therapeutics Al, Corporation Oligomer-guanidine class conjugates
JP5693967B2 (en) 2008-01-25 2015-04-01 ウェルズ ファーゴ バンク ナショナル アソシエイション Oligomer-diarylpiperazine conjugates
EP2254601B1 (en) 2008-02-08 2019-05-29 Nektar Therapeutics Oligomer-cannabinoid conjugates
US8466276B2 (en) * 2008-02-22 2013-06-18 Nektar Therapeutics Oligomer conjugates of heteropentacyclic nucleosides
DK2257311T3 (en) 2008-02-27 2014-06-30 Novo Nordisk As Conjugated Factor VIII Molecules
TWI395593B (en) 2008-03-06 2013-05-11 Halozyme Inc In vivo temporal control of activatable matrix-degrading enzymes
US20090226531A1 (en) * 2008-03-07 2009-09-10 Allergan, Inc. Methods and composition for intraocular delivery of therapeutic sirna
US9006219B2 (en) 2008-03-12 2015-04-14 Nektar Therapeutics Oligomer-foscarnet conjugates
WO2009114151A1 (en) * 2008-03-12 2009-09-17 Nektar Therapeutics Oligomer-amino acid and olgomer-atazanavir conjugates
US20110059921A1 (en) * 2008-03-27 2011-03-10 Ektar Therapeutics Oligomer-Nitrogenous Base Conjugates
MX2010010313A (en) * 2008-04-03 2010-11-05 Hoffmann La Roche Pegylated insulin-like-growth-factor assay.
KR101273187B1 (en) * 2008-04-03 2013-06-17 에프. 호프만-라 로슈 아게 Use of pegylated igf-i variants for the treatment of neuromuscular disorders
KR101680385B1 (en) 2008-04-11 2016-11-28 넥타르 테라퓨틱스 Oligomer-aryloxy-substituted propanamine conjugates
AU2009234266B2 (en) 2008-04-11 2015-08-06 Tekmira Pharmaceuticals Corporation Site-specific delivery of nucleic acids by combining targeting ligands with endosomolytic components
WO2009128917A2 (en) 2008-04-14 2009-10-22 Halozyme, Inc. Modified hyaluronidases and uses in treating hyaluronan-associated diseases and conditions
WO2009131695A1 (en) 2008-04-25 2009-10-29 Nektar Therapeutics Oligomer-bis-chromonyl compound conjugates
TWI394580B (en) 2008-04-28 2013-05-01 Halozyme Inc Super fast-acting insulin compositions
US11033631B2 (en) * 2008-06-09 2021-06-15 Nektar Therapeutics Methods of treating CYP2D6 alternative metabolizers
US8815818B2 (en) 2008-07-18 2014-08-26 Rxi Pharmaceuticals Corporation Phagocytic cell delivery of RNAI
CN102159250B (en) 2008-08-11 2014-08-06 尼克塔治疗公司 Multi-arm polymeric alkanoate conjugates
JP5827123B2 (en) 2008-09-16 2015-12-02 ウェルズ ファーゴ バンク ナショナル アソシエイション PEGylated opioids with low potential for abuse
WO2010033219A2 (en) 2008-09-17 2010-03-25 Nektar Therapeutics Protease inhibitors having enhanced features
US20110195912A1 (en) * 2008-09-17 2011-08-11 Nektar Therapeutics Oligomer-Protease Inhibitor Conjugates
AU2009292643B2 (en) * 2008-09-19 2016-02-18 Nektar Therapeutics Polymer conjugates of therapeutic peptides
WO2010033205A1 (en) * 2008-09-19 2010-03-25 Nektar Therapeutics Polymer conjugates of v681-like peptides
US20110171165A1 (en) * 2008-09-19 2011-07-14 Nektar Therapeutics Polymer conjugates of opioid growth factor peptides
US20110171162A1 (en) * 2008-09-19 2011-07-14 Nektar Therapeutics Polymer conjugates of thymosin alpha 1 peptides
US20110165112A1 (en) * 2008-09-19 2011-07-07 Nektar Therapeutics Polymer conjugates of c-peptides
WO2010033222A2 (en) * 2008-09-19 2010-03-25 Netkar Therapeutics Polymer conjugates of ziconotide peptides
US20110171160A1 (en) * 2008-09-19 2011-07-14 Nektar Therapeutics Polymer conjugates of kiss1 peptides
US20110171161A1 (en) * 2008-09-19 2011-07-14 Nektar Therapeutics Polymer conjugates of protegrin peptides
WO2010042145A1 (en) * 2008-09-19 2010-04-15 Nektar Therapeutics Polymer conjugates of glp-2-like peptides
WO2010033216A1 (en) * 2008-09-19 2010-03-25 Nektar Therapeutics Polymer conjugates of nesiritide peptides
WO2010033218A1 (en) * 2008-09-19 2010-03-25 Nektar Therapeutics Polymer conjugates of osteocalcin peptides
WO2010033220A2 (en) * 2008-09-19 2010-03-25 Nektar Therapeutics Modified therapeutics peptides, methods of their preparation and use
AU2009292651A1 (en) * 2008-09-19 2010-03-25 Nektar Therapeutics Polymer conjugates of AOD-like peptides
CN102405286A (en) 2008-09-22 2012-04-04 阿克赛医药公司 Reduced size self-delivering rnai compounds
JP2012513953A (en) 2008-09-23 2012-06-21 アルニラム ファーマスーティカルズ インコーポレイテッド Chemical modification of monomers and oligonucleotides using cycloaddition
WO2010039985A1 (en) 2008-10-01 2010-04-08 Quintessence Biosciences, Inc. Therapeutic Ribonucleases
KR20230147211A (en) 2008-11-10 2023-10-20 알닐람 파마슈티칼스 인코포레이티드 Novel lipids and compositions for the delivery of therapeutics
RS58728B1 (en) 2008-12-09 2019-06-28 Halozyme Inc Extended soluble ph20 polypeptides and uses thereof
WO2010078536A1 (en) 2009-01-05 2010-07-08 Rxi Pharmaceuticals Corporation Inhibition of pcsk9 through rnai
WO2010080833A1 (en) * 2009-01-06 2010-07-15 Dyax Corp. Treatment of mucositis with kallikrein inhibitors
AU2010208274B2 (en) 2009-01-28 2015-11-05 Nektar Therapeutics Oligomer-phenothiazine conjugates
EP2400989B1 (en) 2009-02-24 2016-08-10 Nektar Therapeutics Gabapentin-peg conjugates
US9688631B2 (en) * 2009-03-02 2017-06-27 Isp Investments Llc Thermosetting ring-opening metathesis polymerization materials with thermally degradable linkages
US8975389B2 (en) 2009-03-02 2015-03-10 Alnylam Pharmaceuticals, Inc. Nucleic acid chemical modifications
WO2010104883A1 (en) * 2009-03-09 2010-09-16 Molecular Express, Inc. Methods and compositions for liposomal formulation of antigens and uses thereof
WO2010120386A1 (en) 2009-04-17 2010-10-21 Nektar Therapeutics Oligomer-protein tyrosine kinase inhibitor conjugates
WO2010120388A1 (en) 2009-04-17 2010-10-21 Nektar Therapeutics Oligomer-protein tyrosine kinase inhibitor conjugates
CN101870767B (en) * 2009-04-24 2012-09-12 石药集团中奇制药技术(石家庄)有限公司 Preparation method of pegylated 1,2-bialiphatic acyl phosphatidylethanolamine
EP2427233B1 (en) * 2009-05-04 2016-12-21 Incept Llc Biomaterials for track and puncture closure
EP2430003B1 (en) 2009-05-13 2019-02-27 Nektar Therapeutics Oligomer-containing substituted aromatic triazine compounds
WO2010132693A2 (en) 2009-05-13 2010-11-18 Nektar Therapeutics Oligomer-containing pyrrolidine compounds
AR077041A1 (en) 2009-06-09 2011-07-27 Prolong Pharmaceuticals Inc HEMOGLOBIN COMPOSITIONS. METHODS
NZ622843A (en) 2009-06-10 2015-10-30 Tekmira Pharmaceuticals Corp Improved lipid formulation
EP2440249A2 (en) 2009-06-12 2012-04-18 Nektar Therapeutics Covalent conjugates comprising a protease inhibitor, a water-soluble, non-peptidic oligomer and a lipophilic moiety
SG176947A1 (en) 2009-07-03 2012-01-30 Avipep Pty Ltd Immuno-conjugates and methods for producing them
WO2011003633A1 (en) 2009-07-06 2011-01-13 Alize Pharma Ii Pegylated l-asparaginase
HRP20171180T4 (en) * 2009-07-06 2023-06-09 Jazz Pharmaceuticals Ii Sas Pegylated l-asparaginase
US20110009628A1 (en) * 2009-07-08 2011-01-13 Haiyan Liu Compounds and Compositions for Modulating Lipid Levels and Methods of Preparing Same
AU2010276244B2 (en) 2009-07-21 2015-08-13 Nektar Therapeutics Oligomer-opioid agonist conjugates
BR112012002709A2 (en) 2009-08-05 2019-04-30 Pieris Ag controlled release formulations of lipocalin muteins
MX2012003282A (en) 2009-09-17 2012-04-30 Baxter Healthcare Sa Stable co-formulation of hyaluronidase and immunoglobulin, and methods of use thereof.
WO2011035065A1 (en) 2009-09-17 2011-03-24 Nektar Therapeutics Monoconjugated chitosans as delivery agents for small interfering nucleic acids
EP2482855B1 (en) 2009-09-29 2017-12-27 Nektar Therapeutics Oligomer-calcimimetic conjugates and related compounds
US8722732B2 (en) 2009-09-29 2014-05-13 Nektar Therapeutics Oligomer-calcimimetic conjugates and related compounds
US9315860B2 (en) 2009-10-26 2016-04-19 Genovoxx Gmbh Conjugates of nucleotides and method for the application thereof
WO2011071860A2 (en) 2009-12-07 2011-06-16 Alnylam Pharmaceuticals, Inc. Compositions for nucleic acid delivery
US20130017223A1 (en) 2009-12-18 2013-01-17 The University Of British Columbia Methods and compositions for delivery of nucleic acids
KR101961495B1 (en) 2009-12-23 2019-03-22 아비펩 피티와이 리미티트 Immuno-conjugates and methods for producing them 2
US20110152188A1 (en) * 2009-12-23 2011-06-23 Hanns-Christian Mahler Pharmaceutical compositions of igf/i proteins
PT3459564T (en) * 2010-01-06 2022-01-31 Takeda Pharmaceuticals Co Plasma kallikrein binding proteins
US20130023553A1 (en) 2010-01-12 2013-01-24 Nektar Therapeutics Pegylated opioids with low potential for abuse and side effects
WO2011091050A1 (en) 2010-01-19 2011-07-28 Nektar Therapeutics Oligomer-tricyclic conjugates
WO2011100131A2 (en) 2010-01-28 2011-08-18 Alnylam Pharmacuticals, Inc. Monomers and oligonucleotides comprising cycloaddition adduct(s)
WO2011094580A2 (en) 2010-01-28 2011-08-04 Alnylam Pharmaceuticals, Inc. Chelated copper for use in the preparation of conjugated oligonucleotides
WO2011103559A1 (en) 2010-02-22 2011-08-25 Nektar Therapeutics Oligomer modified diaromatic substituted compounds
US20110224383A1 (en) * 2010-03-11 2011-09-15 Intezyne Technologies, Inc. Poly(ethylene glycol) derivatives for metal-free click chemistry
US9102938B2 (en) 2010-04-01 2015-08-11 Alnylam Pharmaceuticals, Inc. 2′ and 5′ modified monomers and oligonucleotides
WO2011133868A2 (en) 2010-04-22 2011-10-27 Alnylam Pharmaceuticals, Inc. Conformationally restricted dinucleotide monomers and oligonucleotides
US10913767B2 (en) 2010-04-22 2021-02-09 Alnylam Pharmaceuticals, Inc. Oligonucleotides comprising acyclic and abasic nucleosides and analogs
WO2011133871A2 (en) 2010-04-22 2011-10-27 Alnylam Pharmaceuticals, Inc. 5'-end derivatives
NZ603399A (en) 2010-05-17 2014-09-26 Cebix Inc Pegylated c-peptide
WO2011163121A1 (en) 2010-06-21 2011-12-29 Alnylam Pharmaceuticals, Inc. Multifunctional copolymers for nucleic acid delivery
ES2661089T3 (en) 2010-07-20 2018-03-27 Halozyme Inc. Methods of treatment or prevention of adverse side effects associated with the administration of an anti-hyaluronan agent
WO2012016184A2 (en) 2010-07-30 2012-02-02 Alnylam Pharmaceuticals, Inc. Methods and compositions for delivery of active agents
WO2012016188A2 (en) 2010-07-30 2012-02-02 Alnylam Pharmaceuticals, Inc. Methods and compositions for delivery of active agents
EP3527220A1 (en) 2010-08-12 2019-08-21 AC Immune S.A. Vaccine engineering
WO2012044992A2 (en) 2010-09-30 2012-04-05 Agency For Science, Technology And Research (A*Star) Methods and reagents for detection and treatment of esophageal metaplasia
WO2012051551A1 (en) 2010-10-15 2012-04-19 Nektar Therapeutics N-optionally substituted aryl-2-oligomer-3-alkoxypropionamides
WO2012054822A1 (en) 2010-10-22 2012-04-26 Nektar Therapeutics Pharmacologically active polymer-glp-1 conjugates
TW201223561A (en) 2010-10-26 2012-06-16 Ac Immune Sa Preparation of an antigenic construct
TWI557135B (en) 2010-11-03 2016-11-11 介控生化科技公司 Modified factor ix polypeptides and uses thereof
US9090535B2 (en) 2010-12-10 2015-07-28 Nektar Therapeutics Hydroxylated tricyclic compounds
WO2012082995A1 (en) 2010-12-15 2012-06-21 Nektar Therapeutics Oligomer-containing hydantoin compounds
WO2012083153A1 (en) 2010-12-16 2012-06-21 Nektar Therapeutics Oligomer-containing apremilast moiety compounds
US20140371258A1 (en) 2010-12-17 2014-12-18 Nektar Therapeutics Water-Soluble Polymer Conjugates of Topotecan
JP5899241B2 (en) 2010-12-21 2016-04-06 ネクター セラピューティクス Multi-arm polymer prodrug conjugates of pemetrexed-based compounds
WO2012088422A1 (en) 2010-12-22 2012-06-28 Nektar Therapeutics Multi-arm polymeric prodrug conjugates of taxane-based compounds
US10894087B2 (en) 2010-12-22 2021-01-19 Nektar Therapeutics Multi-arm polymeric prodrug conjugates of cabazitaxel-based compounds
CN105713092B (en) 2011-01-06 2019-09-10 戴埃克斯有限公司 Blood plasma prekallikrein binding protein
EP3202760B1 (en) 2011-01-11 2019-08-21 Alnylam Pharmaceuticals, Inc. Pegylated lipids and their use for drug delivery
JP2014510045A (en) 2011-02-08 2014-04-24 ハロザイム インコーポレイテッド Hyaluronan degrading enzyme composition and lipid preparation and its use for the treatment of benign prostatic hypertrophy
US9944688B2 (en) * 2011-06-02 2018-04-17 Hanmi Science Co., Ltd. Non-peptidyl polymer-insulin multimer and method for producing the same
CN105797140B (en) 2011-06-17 2020-08-11 哈洛齐梅公司 Stable formulations of hyaluronan degrading enzymes
CA2839512C (en) 2011-06-17 2018-01-02 Halozyme, Inc. Continuous subcutaneous insulin infusion methods with a hyaluronan-degrading enzyme
US9993529B2 (en) 2011-06-17 2018-06-12 Halozyme, Inc. Stable formulations of a hyaluronan-degrading enzyme
US20130071394A1 (en) 2011-09-16 2013-03-21 John K. Troyer Compositions and combinations of organophosphorus bioscavengers and hyaluronan-degrading enzymes, and methods of use
US9701623B2 (en) 2011-09-27 2017-07-11 Alnylam Pharmaceuticals, Inc. Di-aliphatic substituted pegylated lipids
WO2013063155A2 (en) 2011-10-24 2013-05-02 Halozyme, Inc. Companion diagnostic for anti-hyaluronan agent therapy and methods of use thereof
US10525054B2 (en) 2011-11-07 2020-01-07 Inheris Biopharma, Inc. Compositions, dosage forms, and co-administration of an opioid agonist compound and an analgesic compound
US9457024B2 (en) 2011-11-07 2016-10-04 Nektar Therapeutics Compositions, dosage forms, and co-administration of an opioid agonist compound and a non-steroidal anti-inflammatory drug
US8962553B2 (en) 2011-11-17 2015-02-24 Cebix Ab Method of treating a diabetic subject having a microvascular impairment disorder by a pegylated C-peptide
FR2984328B1 (en) 2011-12-20 2016-12-30 Bio-Rad Innovations METHOD FOR DETECTING HEPATITIS C VIRUS INFECTION
AU2012362141B2 (en) 2011-12-30 2017-09-21 Halozyme, Inc. PH20 polypeptide variants, formulations and uses thereof
ES2661575T3 (en) 2012-03-30 2018-04-02 The Board Of Regents Of The University Of Oklahoma Production of high molecular weight heparosan polymers and methods of production and use thereof
WO2013151991A1 (en) 2012-04-02 2013-10-10 Surmodics, Inc. Hydrophilic polymeric coatings for medical articles with visualization moiety
AU2013243873B2 (en) 2012-04-04 2016-08-25 Halozyme, Inc. Combination therapy with an anti - hyaluronan agent and a tumor - targeted taxane
EP2833883A4 (en) 2012-04-06 2015-12-23 Indus Pharmaceuticals Inc Novel compositions of combinations of non-covalent dna binding agents and anti-cancer and/or anti-inflammatory agents and their use in disease treatment
JP6265998B2 (en) 2012-09-17 2018-01-24 ネクター セラピューティクス Oligomer-containing benzamide compounds
WO2014062856A1 (en) 2012-10-16 2014-04-24 Halozyme, Inc. Hypoxia and hyaluronan and markers thereof for diagnosis and monitoring of diseases and conditions and related methods
SG10201804331TA (en) 2012-11-15 2018-07-30 Roche Innovation Ct Copenhagen As Oligonucleotide conjugates
US9580393B2 (en) 2012-12-13 2017-02-28 University Of Kansas 6-substituted quinazolinone inhibitors
ES2927758T3 (en) 2013-03-14 2022-11-10 Alere San Diego Inc 6-Acetylmorphine analogues and procedures for their synthesis and use
CN105209033B (en) * 2013-03-14 2018-09-18 马萨诸塞大学 Inhibit cataract and presbyopic method
WO2014160871A2 (en) 2013-03-27 2014-10-02 The General Hospital Corporation Methods and agents for treating alzheimer's disease
TW201534726A (en) 2013-07-03 2015-09-16 Halozyme Inc Thermally stable PH20 hyaluronidase variants and uses thereof
US9926275B2 (en) 2013-08-22 2018-03-27 Northeastern University Allosteric modulators of the cannabinoid 1 receptor
MX2016004609A (en) 2013-10-15 2016-08-01 Seattle Genetics Inc Pegylated drug-linkers for improved ligand-drug conjugate pharmacokinetics.
EP3078699B1 (en) 2013-12-02 2019-08-21 Jenkem Technology Co. Ltd. (Tianjin) Multi-arm polyethylene glycol-nitrine derivative
CN104774161B (en) * 2014-01-13 2017-08-25 成都福瑞康生物科技有限公司 Polypeptide, protein PEG dressing agent synthetic methods
EP3122782A4 (en) 2014-03-27 2017-09-13 Dyax Corp. Compositions and methods for treatment of diabetic macular edema
US10358636B2 (en) 2014-05-14 2019-07-23 Stealth Biologics, Llc Deimmunized lysostaphin and methods of use
US9611270B2 (en) 2014-08-01 2017-04-04 University Of Kansas Inhibitors of CYP17A1
HUE043847T2 (en) 2014-08-28 2019-09-30 Halozyme Inc Combination therapy with a hyaluronan-degrading enzyme and an immune checkpoint inhibitor
ES2753391T3 (en) 2014-10-14 2020-04-08 Halozyme Inc Adenosine deaminase 2 (ADA2) compositions, variants thereof and methods of use thereof
US10118896B2 (en) 2014-11-26 2018-11-06 University Of Kansas Antagonists of the kappa opioid receptor
WO2016100716A1 (en) 2014-12-18 2016-06-23 Vasant Jadhav Reversirtm compounds
JP6638970B2 (en) 2015-06-30 2020-02-05 株式会社 東北テクノアーチ Method for producing hetero-type monodisperse polyethylene glycol and intermediate for producing hetero-type monodisperse polyethylene glycol
CA2989970A1 (en) 2015-07-17 2017-01-26 Alnylam Pharmaceuticals, Inc. Multi-targeted single entity conjugates
PE20181070A1 (en) 2015-11-13 2018-07-04 Univ Massachusetts BIFUNCTIONAL MOLECULES CONTAINING PEG FOR USE IN THE INHIBITION OF CATARACT AND PRESBYCIA
US11793880B2 (en) 2015-12-04 2023-10-24 Seagen Inc. Conjugates of quaternized tubulysin compounds
WO2017096311A1 (en) 2015-12-04 2017-06-08 Seattle Genetics, Inc. Conjugates of quaternized tubulysin compounds
CN108602893A (en) 2015-12-11 2018-09-28 戴埃克斯有限公司 Inhibitors of plasma kallikrein and its purposes for the treatment of hereditary angioedema breaking-out
WO2017165851A1 (en) 2016-03-25 2017-09-28 Seattle Genetics, Inc. Process for the preparation of pegylated drug-linkers and intermediates thereof
CN107375288B (en) 2016-05-16 2019-08-23 博瑞生物医药(苏州)股份有限公司 The polymerization target anticancer conjugate of multi-arm
AU2017267728B2 (en) 2016-05-18 2021-11-04 Alere San Diego, Inc. 2-ethylidene-1,5-dimethyl-3,3-diphenylpyrrolidine analogs and methods for their synthesis and use
CA3028978A1 (en) 2016-06-23 2017-12-28 Cornell University Double targeted constructs to affect tumor kill
CN115716808A (en) 2016-06-28 2023-02-28 康奈尔大学 18F-labeled triazoles containing PSMA inhibitors
CN107973790A (en) 2016-10-22 2018-05-01 合帕吉恩治疗公司 Heterocyclic FXR conditioning agent
CN108017636A (en) 2016-11-04 2018-05-11 合帕吉恩治疗公司 Nitrogen-containing heterocycle compound as FXR conditioning agents
JP7244987B2 (en) 2016-12-14 2023-03-23 シージェン インコーポレイテッド Multidrug Antibody Drug Conjugates
US10040831B2 (en) 2016-12-19 2018-08-07 Morehouse School Of Medicine Compositions and methods for treating diseases by inhibiting exosome release
US10800817B2 (en) 2016-12-19 2020-10-13 Morehouse School Of Medicine Compositions and methods for treating diseases by inhibiting exosome release
EP3600443A1 (en) 2017-03-24 2020-02-05 Seattle Genetics, Inc. Process for the preparation of glucuronide drug-linkers and intermediates thereof
CA3066915A1 (en) 2017-06-11 2018-12-20 Molecular Express, Inc. Methods and compositions for substance use disorder vaccine formulations and uses thereof
AU2018290281B2 (en) 2017-06-22 2021-12-16 Vertex Pharmaceuticals Incorporated Modified membrane type serine protease 1 (MTSP-1) polypeptides and methods of use
US11491212B1 (en) 2017-09-27 2022-11-08 Catalyst Biosciences, Inc. Subcutaneous administration of modified factor IX polypeptides and treatment of hemophilia B
KR20210018267A (en) 2018-05-07 2021-02-17 알닐람 파마슈티칼스 인코포레이티드 Extrahepatic delivery
WO2019222435A1 (en) 2018-05-16 2019-11-21 Halozyme, Inc. Methods of selecting subjects for combination cancer therapy with a polymer-conjugated soluble ph20
TW202015740A (en) 2018-06-07 2020-05-01 美商西雅圖遺傳學公司 Camptothecin conjugates
AU2019350765A1 (en) 2018-09-28 2021-05-27 Alnylam Pharmaceuticals, Inc. Transthyretin (TTR) iRNA compositions and methods of use thereof for treating or preventing TTR-associated ocular diseases
US11613744B2 (en) 2018-12-28 2023-03-28 Vertex Pharmaceuticals Incorporated Modified urokinase-type plasminogen activator polypeptides and methods of use
KR20210110848A (en) 2018-12-28 2021-09-09 카탈리스트 바이오사이언시즈, 인코포레이티드 Modified urokinase type plasminogen activator polypeptides and methods of use
EP3969052A1 (en) 2019-05-17 2022-03-23 Alnylam Pharmaceuticals Inc. Oral delivery of oligonucleotides
CA3152316A1 (en) 2019-10-04 2021-04-08 Scott C. Jeffrey Camptothecin peptide conjugates
MX2022005220A (en) 2019-11-06 2022-08-11 Alnylam Pharmaceuticals Inc Extrahepatic delivery.
EP4055165A1 (en) 2019-11-06 2022-09-14 Alnylam Pharmaceuticals, Inc. Transthyretin (ttr) irna compositions and methods of use thereof for treating or preventing ttr-associated ocular diseases
WO2021154414A2 (en) 2020-01-29 2021-08-05 Catalyst Biosciences, Inc. Gene therapy for hemophilia b with a chimeric aav capsid vector encoding modified factor ix polypeptides
JP2023520930A (en) 2020-04-10 2023-05-22 シージェン インコーポレイテッド charge diversity linker
KR20230017207A (en) 2020-04-30 2023-02-03 사이로파 비.브이. Anti-CD103 antibody
US20230257745A1 (en) 2020-07-10 2023-08-17 Alnylam Pharmaceuticals, Inc. Circular siRNAs
WO2022147223A2 (en) 2020-12-31 2022-07-07 Alnylam Pharmaceuticals, Inc. 2'-modified nucleoside based oligonucleotide prodrugs
KR20230136130A (en) 2020-12-31 2023-09-26 알닐람 파마슈티칼스 인코포레이티드 Cyclic-disulfide modified phosphate-based oligonucleotide prodrug
TW202246242A (en) 2021-01-15 2022-12-01 美商西健公司 Immunomodulatory antibody-drug conjugates
CN116847886A (en) 2021-02-03 2023-10-03 思进公司 Immunostimulatory compounds and conjugates
US20220313798A1 (en) 2021-03-30 2022-10-06 Jazz Pharmaceuticals Ireland Ltd. Dosing of recombinant l-asparaginase
KR20240015670A (en) 2021-05-28 2024-02-05 씨젠 인크. Anthracycline antibody conjugate
US11180534B1 (en) 2021-06-04 2021-11-23 Morehouse School Of Medicine Compositions and methods for treating SARS-CoV-2 infections
WO2023283403A2 (en) 2021-07-09 2023-01-12 Alnylam Pharmaceuticals, Inc. Bis-rnai compounds for cns delivery
WO2023003922A1 (en) 2021-07-21 2023-01-26 Alnylam Pharmaceuticals, Inc. Metabolic disorder-associated target gene irna compositions and methods of use thereof
WO2023064530A1 (en) 2021-10-15 2023-04-20 Alnylam Pharmaceuticals, Inc. Extra-hepatic delivery irna compositions and methods of use thereof
WO2023083919A1 (en) 2021-11-09 2023-05-19 Tubulis Gmbh Conjugates comprising a phosphorus (v) and a camptothecin moiety
US20230330258A1 (en) 2021-11-09 2023-10-19 Tubulis Gmbh Conjugates comprising a phosphorus (v) and a drug moiety
WO2023178289A2 (en) 2022-03-17 2023-09-21 Seagen Inc. Camptothecin conjugates
WO2023199196A1 (en) 2022-04-12 2023-10-19 Galderma Holding SA Salts for mtor compounds
WO2023215740A1 (en) 2022-05-06 2023-11-09 Seagen Inc. Immunomodulatory antibody-drug conjugates
WO2023220744A2 (en) 2022-05-13 2023-11-16 Alnylam Pharmaceuticals, Inc. Single-stranded loop oligonucleotides
WO2024006999A2 (en) 2022-06-30 2024-01-04 Alnylam Pharmaceuticals, Inc. Cyclic-disulfide modified phosphate based oligonucleotide prodrugs
WO2024015529A2 (en) 2022-07-14 2024-01-18 Jazz Pharmaceuticals Ireland Ltd. Combination therapies involving l-asparaginase
WO2024030577A1 (en) 2022-08-03 2024-02-08 Seagen Inc. Immunostimulatory anti-pd-l1-drug conjugates
EP4321522A1 (en) 2022-08-12 2024-02-14 Seagen Inc. Cytotoxic compounds and conjugates thereof

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0067029A2 (en) * 1981-06-10 1982-12-15 Ajinomoto Co., Inc. Oxygen carrier
US5328955A (en) * 1988-11-21 1994-07-12 Collagen Corporation Collagen-polymer conjugates

Family Cites Families (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE2631656C2 (en) * 1976-07-14 1983-02-10 Boehringer Mannheim Gmbh, 6800 Mannheim Use of a hydroxysuccinimido ester derivative for binding biologically active proteins to one another and / or to liquid or solid carriers
EP0206448B1 (en) * 1985-06-19 1990-11-14 Ajinomoto Co., Inc. Hemoglobin combined with a poly(alkylene oxide)
US4943626A (en) * 1988-07-29 1990-07-24 The Dow Chemical Company Primary polyether active hydrogen compounds which are prepared from linked, protectively initiated polyalkyleneoxides
US5157075A (en) * 1990-05-02 1992-10-20 Taenaka Mining Co., Ltd. Modified melanin
AU657483B2 (en) * 1990-07-20 1995-03-16 Pharmacia Ab Heterobifunctional reagents and conjugates with oxaalkylene units for amphiphilic bridge structures
US5483008A (en) * 1992-02-07 1996-01-09 Research Development Corporation Of Japan Polyether having heterofunctional groups at both ends, process for the preparation thereof and polymerization initiator therefor
IT1256420B (en) * 1992-11-17 1995-12-05 Elisabetta Ranucci MONOFUNCTIONAL LOW N-ACRYLOYLMORPHOLINE POLYMERS EXTREMELY CONJUGATED WITH THE ENZYMES AND DRUGS
US5256819A (en) * 1992-12-24 1993-10-26 Shell Oil Company Preparation of polyoxyalkylene-alpha,omega-dicarboxylic acids
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

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0067029A2 (en) * 1981-06-10 1982-12-15 Ajinomoto Co., Inc. Oxygen carrier
US5328955A (en) * 1988-11-21 1994-07-12 Collagen Corporation Collagen-polymer conjugates

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
SAMUEL ZALIPSKY ET AL: "Succinimidyl Carbonates of Polyethylene Glycol: Useful Reactive Polymers for Preparation of Protein Conjugates", POLYMER PREPRINTS, vol. 31, no. 2, 1990 *
SAMUEL ZALIPSKY: "Synthesis of an End-Group Functionalized Polyethylene Glycol-Lipid Conjugate for Preparation of Polymer-Grafted Liposomes", BIOCONJUGATE CHEM., vol. 4, 1993 *

Cited By (200)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6864327B2 (en) 1997-11-06 2005-03-08 Nektar Therapeutics Al, Corporation Heterobifunctional poly(ethylene glycol) derivatives and methods for their preparation
US6448369B1 (en) 1997-11-06 2002-09-10 Shearwater Corporation Heterobifunctional poly(ethylene glycol) derivatives and methods for their preparation
US10035880B2 (en) 1997-11-06 2018-07-31 Nektar Therapeutics Heterobifunctional poly(ethylene glycol) derivatives and methods for their preparation
US8940801B2 (en) 1997-11-06 2015-01-27 Nektar Therapeutics Heterobifunctional poly(ethylene glycol) derivatives and methods for their preparation
US7642323B2 (en) 1997-11-06 2010-01-05 Nektar Therapeutics Heterobifunctional poly(ethylene glycol) derivatives and methods for their preparation
US6541543B2 (en) 1998-03-12 2003-04-01 Shearwater Corporation Poly (ethylene glycol) derivatives with proximal reactive groups
US7714088B2 (en) 1998-03-12 2010-05-11 Nektar Therapeutics Poly(ethylene glycol) derivatives with proximal reactive groups
EP1411075A3 (en) * 1998-03-12 2004-07-28 Nektar Therapeutics Al, Corporation Method for preparing polymer conjugates
US6437025B1 (en) 1998-03-12 2002-08-20 Shearwater Corporation Poly(ethylene glycol) derivatives with proximal reactive groups
US7528202B2 (en) 1998-03-12 2009-05-05 Nektar Therapeutics Al, Corporation Poly (ethylene glycol) derivatives with proximal reactive groups
US6664331B2 (en) 1998-03-12 2003-12-16 Nektar Therapeutics Al, Corporation Poly(ethylene glycol) derivatives with proximal reactive groups
US7030278B2 (en) 1998-03-12 2006-04-18 Nektar Therapeutics Al, Corporation Polyethylene(Glycol) derivatives with proximal reactive groups
US8003742B2 (en) 1998-03-12 2011-08-23 Nektar Therapeutics Polymer derivatives with proximal reactive groups
US7223803B2 (en) 1998-03-12 2007-05-29 Nektar Therapeutics Al, Corporation Polyethylene (glycol) derivatives with proximal reactive groups
WO1999045964A1 (en) * 1998-03-12 1999-09-16 Shearwater Polymers, Incorporated Poly(ethylene glycol) derivatives with proximal reactive groups
EP1411075A2 (en) * 1998-03-12 2004-04-21 Nektar Therapeutics Al, Corporation Method for preparing polymer conjugates
WO2001026692A1 (en) * 1999-10-08 2001-04-19 Shearwater Corporation Heterobifunctional poly(ethylene glycol) derivatives and methods for their preparation
US8183338B2 (en) 1999-12-22 2012-05-22 Nektar Therapeutics Sterically hindered poly(ethylene glycol) alkanoic acids and derivatives thereof
US6737505B2 (en) 1999-12-22 2004-05-18 Nektar Therapeutics Al, Corporation Sterically hindered poly(ethylene glycol) alkanoic acids and derivatives thereof
US7405266B2 (en) 1999-12-22 2008-07-29 Nektar Therapeutics Al, Corporation Sterically hindered poly(ethylene glycol) alkanoic acids and derivatives thereof
US7960502B2 (en) 1999-12-22 2011-06-14 Nektar Theapeutics Sterically hindered poly(ethylene glycol) alkanoic acids and derivatives thereof
US7205380B2 (en) 1999-12-22 2007-04-17 Nektar Therapeutics Al, Corporation Sterically hindered poly (ethylene glycol) alkanoic acids and derivatives thereof
AU781839B2 (en) * 1999-12-22 2005-06-16 Nektar Therapeutics Sterically hindered derivatives of water soluble polymers
US6495659B2 (en) 1999-12-22 2002-12-17 Shearwater Corporation Sterically hindered poly(ethylene glycol) alkanoic acids and derivatives thereof
WO2001046291A1 (en) * 1999-12-22 2001-06-28 Shearwater Corporation Sterically hindered derivatives of water soluble polymers
US6992168B2 (en) 1999-12-22 2006-01-31 Nektar Therapeutics Al, Corporation Sterically hindered poly(ethylene glycol) alkanoic acids and derivatives thereof
US7674879B2 (en) 1999-12-22 2010-03-09 Nektar Therapeutics Sterically hindered poly(ethylene glycol) alkanoic acids and derivatives thereof
EP2133098A1 (en) 2000-01-10 2009-12-16 Maxygen Holdings Ltd G-CSF conjugates
EP2319541A1 (en) 2000-02-11 2011-05-11 Bayer HealthCare LLC Factor VII or VIIA-like conjugates
EP1982732A2 (en) 2000-02-11 2008-10-22 Maxygen Holdings Ltd. Factor VII or VIIA-like molecules
US7128913B2 (en) 2000-12-20 2006-10-31 Hoffmann-La Roche Inc. Erythropoietin conjugates
WO2002049673A3 (en) * 2000-12-20 2003-01-23 Hoffmann La Roche Conjugates of erythropoietin (pep) with polyethylene glycol (peg)
WO2002049673A2 (en) 2000-12-20 2002-06-27 F. Hoffmann-La Roche Ag Conjugates of erythropoietin (pep) with polyethylene glycol (peg)
EP2080771A2 (en) 2001-02-27 2009-07-22 Maxygen Aps New interferon beta-like molecules
EP1423093A4 (en) * 2001-04-23 2005-11-30 Wisconsin Alumni Res Found Bifunctional-modified hydrogels
US8025901B2 (en) 2001-04-23 2011-09-27 Wisconsin Alumni Research Foundation Bifunctional-modified hydrogels
EP1423093A2 (en) * 2001-04-23 2004-06-02 Wisconsin Alumni Research Foundation Bifunctional-modified hydrogels
US7615593B2 (en) 2001-04-23 2009-11-10 Wisconsin Alumni Research Foundation Bifunctional-modified hydrogels
WO2003005026A3 (en) * 2001-07-02 2003-06-26 Amersham Biosciences Uk Ltd Chemical capture reagent
WO2003005026A2 (en) * 2001-07-02 2003-01-16 Amersham Biosciences Uk Limited Chemical capture reagent
WO2004000366A1 (en) 2002-06-21 2003-12-31 Novo Nordisk Health Care Ag Pegylated factor vii glycoforms
US7504477B2 (en) 2002-07-24 2009-03-17 Hoffmann-La Roche Inc. Polyalkylene glycol acid additives
WO2004012773A1 (en) 2002-07-24 2004-02-12 F. Hoffmann-La Roche Ag Polyalkylene glycol acid additives
US7193031B2 (en) 2002-07-24 2007-03-20 Hoffmann-La Roche Inc. Polyalkylene glycol acid additives
WO2004022629A2 (en) * 2002-09-09 2004-03-18 Nektar Therapeutics Al, Corporation Method for preparing water-soluble polymer derivatives bearing a terminal carboxylic acid
US7569214B2 (en) 2002-09-09 2009-08-04 Nektar Therapeutics Al, Corporation Method for preparing water-soluble polymer derivatives bearing a terminal carboxylic acid
US8784791B2 (en) 2002-09-09 2014-07-22 Nektar Therapeutics Method for preparing water-soluble polymer derivatives bearing an N-succinimidyl ester
US8182801B2 (en) 2002-09-09 2012-05-22 Nektar Therapeutics Method for preparing water-soluble polymer derivatives bearing a terminal carboxylic acid
AU2003270555B2 (en) * 2002-09-09 2009-01-29 Nektar Therapeutics Method for preparing water-soluble polymer derivatives bearing a terminal carboxylic acid
US9045494B2 (en) 2002-09-09 2015-06-02 Nektar Therapeutics Orthoester compound
KR100967334B1 (en) 2002-09-09 2010-07-05 넥타르 테라퓨틱스 Method for preparing water-soluble polymer derivatives bearing a terminal carboxylic acid
US8435504B2 (en) 2002-09-09 2013-05-07 Nektar Therapeutics Method for preparing water-soluble polymer derivatives bearing a terminal carboxylic acid
WO2004022629A3 (en) * 2002-09-09 2004-04-08 Nektar Therapeutics Al Corp Method for preparing water-soluble polymer derivatives bearing a terminal carboxylic acid
WO2004061094A1 (en) 2002-12-30 2004-07-22 Gryphon Therapeutics, Inc. Water-soluble thioester and selenoester compounds and methods for making and using the same
US8034900B2 (en) 2002-12-30 2011-10-11 Amylin Pharmaceuticals, Inc. Water-soluble thioester and selenoester compounds and methods for making and using the same
EP2390262A1 (en) 2003-05-16 2011-11-30 Intermune, Inc. Synthetic chemokine receptor ligands and methods of use thereof
EP2263684A1 (en) 2003-10-10 2010-12-22 Novo Nordisk A/S IL-21 derivatives
WO2005037214A2 (en) 2003-10-14 2005-04-28 Intermune, Inc. Macrocyclic carboxylic acids and acylsulfonamides as inhibitors of hcv replication
EP2407470A2 (en) 2003-10-14 2012-01-18 F. Hoffmann-La Roche Ltd. Macrocyclic carboxylic acids and acylsulfonamides as inhibitors of HCV replication
EP2633866A2 (en) 2003-10-17 2013-09-04 Novo Nordisk A/S Combination therapy
EP2641611A2 (en) 2003-10-17 2013-09-25 Novo Nordisk A/S Combination therapy
US9920106B2 (en) 2003-12-18 2018-03-20 Novo Nordisk A/S GLP-1 compounds
US7608663B2 (en) 2004-01-21 2009-10-27 Nektar Therapeutics Method of preparing propionic acid-terminated polymers
US8097702B2 (en) 2004-02-02 2012-01-17 Ambrx, Inc. Modified human interferon polypeptides with at least one non-naturally encoded amino acid and their uses
US8232371B2 (en) 2004-02-02 2012-07-31 Ambrx, Inc. Modified human interferon polypeptides and their uses
EP2327724A2 (en) 2004-02-02 2011-06-01 Ambrx, Inc. Modified human growth hormone polypeptides and their uses
US8907064B2 (en) 2004-02-02 2014-12-09 Ambrx, Inc. Modified human four helical bundle polypeptides and their uses
US9260472B2 (en) 2004-02-02 2016-02-16 Ambrx, Inc. Modified human four helical bundle polypeptides and their uses
US8906676B2 (en) 2004-02-02 2014-12-09 Ambrx, Inc. Modified human four helical bundle polypeptides and their uses
WO2006009901A2 (en) 2004-06-18 2006-01-26 Ambrx, Inc. Novel antigen-binding polypeptides and their uses
US9175083B2 (en) 2004-06-18 2015-11-03 Ambrx, Inc. Antigen-binding polypeptides and their uses
US8178108B2 (en) 2004-12-22 2012-05-15 Ambrx, Inc. Methods for expression and purification of recombinant human growth hormone
US7838265B2 (en) 2004-12-22 2010-11-23 Ambrx, Inc. Compositions of aminoacyl-tRNA synthetase and uses thereof
US7883866B2 (en) 2004-12-22 2011-02-08 Ambrx, Inc. Compositions of aminoacyl-tRNA synthetase and uses thereof
US7959926B2 (en) 2004-12-22 2011-06-14 Ambrx, Inc. Methods for expression and purification of recombinant human growth hormone mutants
US8178494B2 (en) 2004-12-22 2012-05-15 Ambrx, Inc. Modified human growth hormone formulations with an increased serum half-life
US7846689B2 (en) 2004-12-22 2010-12-07 Ambrx, Inc. Compositions of aminoacyl-tRNA synthetase and uses thereof
EP2284191A2 (en) 2004-12-22 2011-02-16 Ambrx, Inc. Process for the preparation of hGH
US7939496B2 (en) 2004-12-22 2011-05-10 Ambrx, Inc. Modified human growth horomone polypeptides and their uses
US7858344B2 (en) 2004-12-22 2010-12-28 Ambrx, Inc. Compositions of aminoacyl-tRNA synthetase and uses thereof
US7829310B2 (en) 2004-12-22 2010-11-09 Ambrx, Inc. Compositions of aminoacyl-tRNA synthetase and uses thereof
US7816320B2 (en) 2004-12-22 2010-10-19 Ambrx, Inc. Formulations of human growth hormone comprising a non-naturally encoded amino acid at position 35
US8163695B2 (en) 2004-12-22 2012-04-24 Ambrx Formulations of human growth hormone comprising a non-naturally encoded amino acid
US8143216B2 (en) 2004-12-22 2012-03-27 Ambrx, Inc. Modified human growth hormone
US7736872B2 (en) 2004-12-22 2010-06-15 Ambrx, Inc. Compositions of aminoacyl-TRNA synthetase and uses thereof
US8080391B2 (en) 2004-12-22 2011-12-20 Ambrx, Inc. Process of producing non-naturally encoded amino acid containing high conjugated to a water soluble polymer
US7947473B2 (en) 2004-12-22 2011-05-24 Ambrx, Inc. Methods for expression and purification of pegylated recombinant human growth hormone containing a non-naturally encoded keto amino acid
EP2314320A2 (en) 2005-04-05 2011-04-27 Istituto di Richerche di Biologia Molecolare P. Angeletti S.p.A. Method for shielding functional sites or epitopes on proteins
EP2279756A2 (en) 2005-04-05 2011-02-02 Instituto di Ricerche di Biologia Molecolare p Angeletti S.P.A. Method for shielding functional sites or epitopes on proteins
US8093356B2 (en) 2005-06-03 2012-01-10 Ambrx, Inc. Pegylated human interferon polypeptides
EP2360170A2 (en) 2005-06-17 2011-08-24 Novo Nordisk Health Care AG Selective reduction and derivatization of engineered proteins comprinsing at least one non-native cysteine
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
US7897698B2 (en) 2005-08-26 2011-03-01 Novo Nordisk A/S Method of modifying a macromolecular system
EP2045282A1 (en) * 2005-08-26 2009-04-08 Novo Nordisk A/S A method of modifying a macromolecular system
WO2007022780A1 (en) * 2005-08-26 2007-03-01 Novo Nordisk A/S A method of modifying a macromolecular system
US9488660B2 (en) 2005-11-16 2016-11-08 Ambrx, Inc. Methods and compositions comprising non-natural amino acids
EP2093235A1 (en) 2006-02-08 2009-08-26 Alios Biopharma Inc. Hyperglycosylated variants of interferon alfacon-1
EP2213733A2 (en) 2006-05-24 2010-08-04 Novo Nordisk Health Care AG Factor IX analogues having prolonged in vivo half life
US8420792B2 (en) 2006-09-08 2013-04-16 Ambrx, Inc. Suppressor tRNA transcription in vertebrate cells
US9133495B2 (en) 2006-09-08 2015-09-15 Ambrx, Inc. Hybrid suppressor tRNA for vertebrate cells
WO2008030558A2 (en) 2006-09-08 2008-03-13 Ambrx, Inc. Modified human plasma polypeptide or fc scaffolds and their uses
US8053560B2 (en) 2006-09-08 2011-11-08 Ambrx, Inc. Modified human plasma polypeptide or Fc scaffolds and their uses
US8022186B2 (en) 2006-09-08 2011-09-20 Ambrx, Inc. Modified human plasma polypeptide or Fc scaffolds and their uses
US8618257B2 (en) 2006-09-08 2013-12-31 Ambrx, Inc. Modified human plasma polypeptide or Fc scaffolds and their uses
US7919591B2 (en) 2006-09-08 2011-04-05 Ambrx, Inc. Modified human plasma polypeptide or Fc scaffolds and their uses
EP2548967A2 (en) 2006-09-21 2013-01-23 The Regents of The University of California Aldehyde tags, uses thereof in site-specific protein modification
US8383365B2 (en) 2007-03-30 2013-02-26 Ambrx, Inc. Methods of making FGF-21 mutants comprising non-naturally encoded phenylalanine derivatives
US9079971B2 (en) 2007-03-30 2015-07-14 Ambrx, Inc. Modified FGF-21 polypeptides comprising non-naturally occurring amino acids
US10377805B2 (en) 2007-03-30 2019-08-13 Ambrx, Inc. Modified FGF-21 polypeptides comprising non-naturally encoding amino acids and their uses
US9517273B2 (en) 2007-03-30 2016-12-13 Ambrx, Inc. Methods of treatment using modified FGF-21 polypeptides comprising non-naturally occurring amino acids
US8012931B2 (en) 2007-03-30 2011-09-06 Ambrx, Inc. Modified FGF-21 polypeptides and their uses
US9975936B2 (en) 2007-03-30 2018-05-22 Ambrx, Inc. Nucleic acids encoding modified FGF-21 polypeptides comprising non-naturally occurring amino acids
US10961291B2 (en) 2007-03-30 2021-03-30 Ambrx, Inc. Modified FGF-21 polypeptides and their uses
US8114630B2 (en) 2007-05-02 2012-02-14 Ambrx, Inc. Modified interferon beta polypeptides and their uses
EP2930182A1 (en) 2007-11-20 2015-10-14 Ambrx, Inc. Modified insulin polypeptides and their uses
WO2009067636A2 (en) 2007-11-20 2009-05-28 Ambrx, Inc. Modified insulin polypeptides and their uses
US8946148B2 (en) 2007-11-20 2015-02-03 Ambrx, Inc. Modified insulin polypeptides and their uses
WO2009093713A1 (en) * 2008-01-25 2009-07-30 Ebara Corporation Peg-modified hydroxyapatite, pharmaceutical preparation containing the same as base material, and method for production of the same
EP3103880A1 (en) 2008-02-08 2016-12-14 Ambrx, Inc. Modified leptin polypeptides and their uses
US9938333B2 (en) 2008-02-08 2018-04-10 Ambrx, Inc. Modified leptin polypeptides and their uses
WO2010007626A1 (en) * 2008-07-14 2010-01-21 Biocon Limited A method of synthesizing a substantially monodispersed mixture of oligomers
US9040723B2 (en) 2008-07-14 2015-05-26 Biocon Limited Method of synthesizing a substantially monodispersed mixture of oligomers
US10138283B2 (en) 2008-07-23 2018-11-27 Ambrx, Inc. Modified bovine G-CSF polypeptides and their uses
EP3225248A1 (en) 2008-07-23 2017-10-04 Ambrx, Inc. Modified bovine g-csf polypeptides and their uses
WO2010011735A2 (en) 2008-07-23 2010-01-28 Ambrx, Inc. Modified bovine g-csf polypeptides and their uses
US10428333B2 (en) 2008-09-26 2019-10-01 Ambrx Inc. Non-natural amino acid replication-dependent microorganisms and vaccines
EP3216800A1 (en) 2008-09-26 2017-09-13 Ambrx, Inc. Modified animal erythropoietin polypeptides and their uses
US9121024B2 (en) 2008-09-26 2015-09-01 Ambrx, Inc. Non-natural amino acid replication-dependent microorganisms and vaccines
US9121025B2 (en) 2008-09-26 2015-09-01 Ambrx, Inc. Non-natural amino acid replication-dependent microorganisms and vaccines
US8569233B2 (en) 2008-09-26 2013-10-29 Eli Lilly And Company Modified animal erythropoietin polypeptides and their uses
US9156899B2 (en) 2008-09-26 2015-10-13 Eli Lilly And Company Modified animal erythropoietin polypeptides and their uses
US9644014B2 (en) 2008-09-26 2017-05-09 Ambrx, Inc. Modified animal erythropoietin polypeptides and their uses
US8278418B2 (en) 2008-09-26 2012-10-02 Ambrx, Inc. Modified animal erythropoietin polypeptides and their uses
EP2805965A1 (en) 2009-12-21 2014-11-26 Ambrx, Inc. Modified porcine somatotropin polypeptides and their uses
EP2805964A1 (en) 2009-12-21 2014-11-26 Ambrx, Inc. Modified bovine somatotropin polypeptides and their uses
WO2011107591A1 (en) 2010-03-05 2011-09-09 Rigshospitalet Chimeric inhibitor molecules of complement activation
EP3815708A1 (en) 2010-03-05 2021-05-05 Omeros Corporation Chimeric inhibitor molecules of complement activation
WO2011143274A1 (en) 2010-05-10 2011-11-17 Perseid Therapeutics Polypeptide inhibitors of vla4
US10253083B2 (en) 2010-08-17 2019-04-09 Ambrx, Inc. Therapeutic uses of modified relaxin polypeptides
EP4302783A2 (en) 2010-08-17 2024-01-10 Ambrx, Inc. Modified relaxin polypeptides and their uses
US9452222B2 (en) 2010-08-17 2016-09-27 Ambrx, Inc. Nucleic acids encoding modified relaxin polypeptides
US11786578B2 (en) 2010-08-17 2023-10-17 Ambrx, Inc. Modified relaxin polypeptides and their uses
US8735539B2 (en) 2010-08-17 2014-05-27 Ambrx, Inc. Relaxin polypeptides comprising non-naturally encoded amino acids
WO2012024452A2 (en) 2010-08-17 2012-02-23 Ambrx, Inc. Modified relaxin polypeptides and their uses
US11439710B2 (en) 2010-08-17 2022-09-13 Ambrx, Inc. Nucleic acids encoding modified relaxin polypeptides
US9567386B2 (en) 2010-08-17 2017-02-14 Ambrx, Inc. Therapeutic uses of modified relaxin polypeptides
US9962450B2 (en) 2010-08-17 2018-05-08 Ambrx, Inc. Method of treating heart failure with modified relaxin polypeptides
US10702588B2 (en) 2010-08-17 2020-07-07 Ambrx, Inc. Modified relaxin polypeptides comprising a non-naturally encoded amino acid in the A chain
US10751391B2 (en) 2010-08-17 2020-08-25 Ambrx, Inc. Methods of treatment using modified relaxin polypeptides comprising a non-naturally encoded amino acid
US11311605B2 (en) 2010-08-17 2022-04-26 Ambrx, Inc. Methods of treating heart failure and fibrotic disorders using modified relaxin polypeptides
US11273202B2 (en) 2010-09-23 2022-03-15 Elanco Us Inc. Formulations for bovine granulocyte colony stimulating factor and variants thereof
WO2013004607A1 (en) 2011-07-01 2013-01-10 Bayer Intellectual Property Gmbh Relaxin fusion polypeptides and uses thereof
US9382305B2 (en) 2011-07-01 2016-07-05 Bayer Intellectual Property Gmbh Relaxin fusion polypeptides and uses thereof
EP3088005A1 (en) 2011-07-05 2016-11-02 biOasis Technologies Inc P97-antibody conjugates
WO2013006706A1 (en) 2011-07-05 2013-01-10 Bioasis Technologies Inc. P97-antibody conjugates and methods of use
EP3505534A1 (en) 2012-06-08 2019-07-03 Sutro Biopharma, Inc. Antibodies comprising sitespecific nonnatural amino acid residues, methods of their preparation and methods of their use
WO2013185115A1 (en) 2012-06-08 2013-12-12 Sutro Biopharma, Inc. Antibodies comprising site-specific non-natural amino acid residues, methods of their preparation and methods of their use
WO2013189745A2 (en) * 2012-06-18 2013-12-27 Basf Se Agroformulations containing a lactone based alkoxylate
US9591854B2 (en) 2012-06-18 2017-03-14 Basf Se Agroformulations containing a lactone based alkoxylate
WO2013189745A3 (en) * 2012-06-18 2014-02-20 Basf Se Agroformulations containing a lactone based alkoxylate
EP3135690A1 (en) 2012-06-26 2017-03-01 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
WO2014022515A1 (en) 2012-07-31 2014-02-06 Bioasis Technologies, Inc. Dephosphorylated lysosomal storage disease proteins and methods of use thereof
EP3584255A1 (en) 2012-08-31 2019-12-25 Sutro Biopharma, Inc. Modified amino acids comprising an azido group
WO2014036492A1 (en) 2012-08-31 2014-03-06 Sutro Biopharma, Inc. Modified amino acids comprising an azido group
EP4074728A1 (en) 2012-08-31 2022-10-19 Sutro Biopharma, Inc. Modified peptides comprising an azido group
US9579390B2 (en) 2012-11-12 2017-02-28 Redwood Bioscience, Inc. Compounds and methods for producing a conjugate
US9833515B2 (en) 2012-11-16 2017-12-05 Redwood Bioscience, Inc. Hydrazinyl-indole compounds and methods for producing a conjugate
US11426465B2 (en) 2012-11-16 2022-08-30 Redwiid Bioscience, Inc. Hydrazinyl-indole compounds and methods for producing a conjugate
US9605078B2 (en) 2012-11-16 2017-03-28 The Regents Of The University Of California Pictet-Spengler ligation for protein chemical modification
US10314919B2 (en) 2012-11-16 2019-06-11 Redwood Bioscience, Inc. Hydrazinyl-indole compounds and methods for producing a conjugate
US9310374B2 (en) 2012-11-16 2016-04-12 Redwood Bioscience, Inc. Hydrazinyl-indole compounds and methods for producing a conjugate
US10888623B2 (en) 2012-11-16 2021-01-12 Redwood Bioscience, Inc. Hydrazinyl-indole compounds and methods for producing a conjugate
WO2014160438A1 (en) 2013-03-13 2014-10-02 Bioasis Technologies Inc. Fragments of p97 and uses thereof
WO2015006555A2 (en) 2013-07-10 2015-01-15 Sutro Biopharma, Inc. Antibodies comprising multiple site-specific non-natural amino acid residues, methods of their preparation and methods of their use
EP3336103A1 (en) 2013-07-10 2018-06-20 Sutro Biopharma, Inc. Antibodies comprising multiple site-specific non-natural amino acid residues, methods of their preparation and methods of their use
WO2015031673A2 (en) 2013-08-28 2015-03-05 Bioasis Technologies Inc. Cns-targeted conjugates having modified fc regions and methods of use thereof
WO2015054658A1 (en) 2013-10-11 2015-04-16 Sutro Biopharma, Inc. Modified amino acids comprising tetrazine functional groups, methods of preparation, and methods of their use
WO2015081282A1 (en) 2013-11-27 2015-06-04 Redwood Bioscience, Inc. Hydrazinyl-pyrrolo compounds and methods for producing a conjugate
US11248031B2 (en) 2014-10-24 2022-02-15 Bristol-Myers Squibb Company Methods of treating diseases associated with fibrosis using modified FGF-21 polypeptides
US9434778B2 (en) 2014-10-24 2016-09-06 Bristol-Myers Squibb Company Modified FGF-21 polypeptides comprising an internal deletion and uses thereof
US10189883B2 (en) 2014-10-24 2019-01-29 Bristol-Myers Squibb Company Therapeutic uses of modified FGF-21 polypeptides
US9631004B2 (en) 2014-10-24 2017-04-25 Bristol-Myers Squibb Company Modified FGF-21 polypeptides comprising an internal deletion and uses thereof
US10377806B2 (en) 2014-10-24 2019-08-13 Bristol-Myers Squibb Company Methods of treating diseases associated with fibrosis using modified FGF-21 polypeptides and uses thereof
US10266578B2 (en) 2017-02-08 2019-04-23 Bristol-Myers Squibb Company Modified relaxin polypeptides comprising a pharmacokinetic enhancer and uses thereof
US11364281B2 (en) 2017-02-08 2022-06-21 Bristol-Myers Squibb Company Modified relaxin polypeptides comprising a pharmacokinetic enhancer and pharmaceutical compositions thereof
US11185570B2 (en) 2017-02-08 2021-11-30 Bristol-Myers Squibb Company Method of treating cardiovascular disease and heart failure with modified relaxin polypeptides
WO2019133399A1 (en) 2017-12-26 2019-07-04 Becton, Dickinson And Company Deep ultraviolet-excitable water-solvated polymeric dyes
WO2019191482A1 (en) 2018-03-30 2019-10-03 Becton, Dickinson And Company Water-soluble polymeric dyes having pendant chromophores
WO2020023300A1 (en) 2018-07-22 2020-01-30 Bioasis Technologies, Inc. Treatment of lymmphatic metastases
WO2020056066A1 (en) 2018-09-11 2020-03-19 Ambrx, Inc. Interleukin-2 polypeptide conjugates and their uses
WO2020082057A1 (en) 2018-10-19 2020-04-23 Ambrx, Inc. Interleukin-10 polypeptide conjugates, dimers thereof, and their uses
WO2020168017A1 (en) 2019-02-12 2020-08-20 Ambrx, Inc. Compositions containing, methods and uses of antibody-tlr agonist conjugates
WO2021183832A1 (en) 2020-03-11 2021-09-16 Ambrx, Inc. Interleukin-2 polypeptide conjugates and methods of use thereof
WO2021236526A1 (en) 2020-05-18 2021-11-25 Bioasis Technologies, Inc. Compositions and methods for treating lewy body dementia
WO2021255524A1 (en) 2020-06-17 2021-12-23 Bioasis Technologies, Inc. Compositions and methods for treating frontotemporal dementia
WO2022040596A1 (en) 2020-08-20 2022-02-24 Ambrx, Inc. Antibody-tlr agonist conjugates, methods and uses thereof
WO2022212899A1 (en) 2021-04-03 2022-10-06 Ambrx, Inc. Anti-her2 antibody-drug conjugates and uses thereof
EP4155349A1 (en) 2021-09-24 2023-03-29 Becton, Dickinson and Company Water-soluble yellow green absorbing dyes
WO2024007016A2 (en) 2022-07-01 2024-01-04 Beckman Coulter, Inc. Novel fluorescent dyes and polymers from dihydrophenanthrene derivatives
WO2024044327A1 (en) 2022-08-26 2024-02-29 Beckman Coulter, Inc. Dhnt monomers and polymer dyes with modified photophysical properties

Also Published As

Publication number Publication date
US5672662A (en) 1997-09-30
AU6345796A (en) 1997-02-10

Similar Documents

Publication Publication Date Title
US5672662A (en) Poly(ethylene glycol) and related polymers monosubstituted with propionic or butanoic acids and functional derivatives thereof for biotechnical applications
KR100948532B1 (en) Branched polymers and their conjugates
US10385165B2 (en) N-maleimidyl polymer derivatives
KR100499655B1 (en) Heterobifunctional poly(ethylene glycol) derivatives and methods for their preparation
US5990237A (en) Poly(ethylene glycol) aldehyde hydrates and related polymers and applications in modifying amines
US7504477B2 (en) Polyalkylene glycol acid additives
US8183338B2 (en) Sterically hindered poly(ethylene glycol) alkanoic acids and derivatives thereof
US5414135A (en) Vinyl sulfone coupling of polyoxyalkylenes to proteins
US8299173B2 (en) Method involving 1-benzotriazolyl carbonate esters of polymers
US20040115165A1 (en) Bifunctional polyethylene glycol derivatives
US20060155059A1 (en) Polyethylene (glycol) derivatives with proximal reactive groups
WO1997014740A1 (en) Discrete-length polyethylene glycols
KR20040096016A (en) Novel Preparation method of PEG-maleimide PEG derivatives
JPH10505088A (en) Polysaccharides, polypeptides (proteins) and electrophilic polyethylene oxides for surface modification
US20100063328A1 (en) Heterobifunctional poly(ethylene glycol) derivatives and methods for their preparation
US6916962B2 (en) Monofunctional polyethylene glycol aldehydes
US6956135B2 (en) Monofunctional polyethylene glycol aldehydes
US20030153694A1 (en) Novel monofunctional polyethylene glycol aldehydes
US7041855B2 (en) Monofunctional polyethylene glycol aldehydes
KR20220069994A (en) Asymmetric branched degradable polyethylene glycol derivatives
Braunová et al. Degradation behavior of poly (ethylene glycol) diblock and multiblock polymers with hydrolytically degradable ester linkages
US20070141021A1 (en) Methylmaleimidyl polymer derivatives

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

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

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): KE LS MW SD SZ UG AT BE CH DE DK ES FI FR GB GR IE IT LU MC NL PT SE

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

Ref country code: DE

Ref legal event code: 8642

122 Ep: pct application non-entry in european phase
NENP Non-entry into the national phase

Ref country code: CA