CA1217718A - Plasminogen activator derivatives - Google Patents

Plasminogen activator derivatives

Info

Publication number
CA1217718A
CA1217718A CA000443230A CA443230A CA1217718A CA 1217718 A CA1217718 A CA 1217718A CA 000443230 A CA000443230 A CA 000443230A CA 443230 A CA443230 A CA 443230A CA 1217718 A CA1217718 A CA 1217718A
Authority
CA
Canada
Prior art keywords
urokinase
peg
molecular weight
triazine
polyethylene glycol
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Expired
Application number
CA000443230A
Other languages
French (fr)
Inventor
Kimihiro Shimizu
Tsuguji Nakahara
Taketoshi Kinoshita
Jun Takatsuka
Michiko Igarashi
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Nippon Chemiphar Co Ltd
Original Assignee
Nippon Chemiphar Co Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from JP56172908A external-priority patent/JPS5896026A/en
Priority to JP56172908A priority Critical patent/JPS5896026A/en
Priority to BR8206347A priority patent/BR8206347A/en
Priority to US06/437,009 priority patent/US4495285A/en
Priority to CH6304/82A priority patent/CH658669A5/en
Priority to CA000414556A priority patent/CA1203764A/en
Priority to DE19823240174 priority patent/DE3240174A1/en
Priority to FR8218223A priority patent/FR2515684B1/en
Priority to GB08230987A priority patent/GB2110219B/en
Priority to SE8206173A priority patent/SE457800B/en
Priority to US06/546,590 priority patent/US4640835A/en
Application filed by Nippon Chemiphar Co Ltd filed Critical Nippon Chemiphar Co Ltd
Priority to CA000443230A priority patent/CA1217718A/en
Publication of CA1217718A publication Critical patent/CA1217718A/en
Application granted granted Critical
Expired legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/56Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule
    • A61K47/59Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyureas or polyurethanes
    • A61K47/60Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyureas or polyurethanes the organic macromolecular compound being a polyoxyalkylene oligomer, polymer or dendrimer, e.g. PEG, PPG, PEO or polyglycerol
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/14Hydrolases (3)
    • C12N9/48Hydrolases (3) acting on peptide bonds (3.4)
    • C12N9/50Proteinases, e.g. Endopeptidases (3.4.21-3.4.25)
    • C12N9/64Proteinases, e.g. Endopeptidases (3.4.21-3.4.25) derived from animal tissue
    • C12N9/6421Proteinases, e.g. Endopeptidases (3.4.21-3.4.25) derived from animal tissue from mammals
    • C12N9/6424Serine endopeptidases (3.4.21)
    • C12N9/6456Plasminogen activators
    • C12N9/6462Plasminogen activators u-Plasminogen activator (3.4.21.73), i.e. urokinase
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/96Stabilising an enzyme by forming an adduct or a composition; Forming enzyme conjugates
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y304/00Hydrolases acting on peptide bonds, i.e. peptidases (3.4)
    • C12Y304/21Serine endopeptidases (3.4.21)
    • C12Y304/21073Serine endopeptidases (3.4.21) u-Plasminogen activator (3.4.21.73), i.e. urokinase
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides

Abstract

ABSTRACT OF THE DISCLOSURE
Derivatives of a nonimmunogenic plasminogen activator which comprises at least one polyalkylene glycol group chemically bonded with at least one coupling agent to amino acid side chains of said plasminogen activator, wherein said polyalkylene glycol has a molecular weight of about 200-20,000 and is unsubstituted or is substituted with one or more alkyl, alkoxy or alkanoyl groups or a mixture thereof.
The plasminogen activator derivatives have an extended circulating life in the mammalian bloodstream and also inhibit the formation of thrombus in the same.

Description

~ ~ ~Lt ,~7.~"~

BACKGROUND OF THE INVENTION
Field of the Invention ~ his invention relates to derivatives of human-originated non-immunogenic plasminogen activators, and more particularly to such a derivative comprising at least one polyalkylene glycol attached with at least one coupling agent to amino acid side chains of a plasminogen activator of the type described above, the polyalkylene glycol having a molecular weight in the range of 200 -10 20,000 and optionally containing one or more alkyl, alkoxy and/oralkanoyl groups as substituents. Further, the inveniion is concerned with a process for producing such deri-vativesand with a thrombolytic agent containing such derivativeS .
Description of the Prior Art It is known that human tissues contain a variety of substances which activate plasminogen into a fibrinoly-tic enzyme or plasmin. Among such known substances, 'he most representative is a plasminogen activator, i.e.
20 urokinase, which is formed in the kidney tissue and excre~d into urine. Urokinase may be obtained by isolation and purification from human urine, tissue culture or genetic engineering. As fibrinolytic enzyme activators which hav~
nowadays found widespread commercial utility, there exist proteins originated from hemolytic Streptococcus and urokinase which is an enzyme originated from human urine. In view of its immug~nic behavior towards humans,urokinase re~ulting from such source is favorably employed for clinical application.

:~`

7~ ;
-2-Urokinase originated from human urine is believed to con-tain both high molecular weight urokinase (molecular weight: 54,000) and low molecular weight urokinase (molecu-lar weight: 33,000). Uroklnase has been used, in recent years, as a thrombolytic agent or an adiuvant for carcino-static substances, and its consumption for clinical application`is increasing year by year.
However, urokinase is unstable under certain conditions since it is an enzyme and loses its enzymatic activity, for example, in the course of extraction, isolation and purification from a urokinase-bearing raw material, for example, urine; during the lyophilization processing in preparing dosable formulations; during tne heat treatment for deactivating viruses, or when it is placed in a diluted state in a dripping bottle and kept for a prolonged time period in such a diluted state at room temperature for clinical application. This physically unstable nature of urokinase has created a serious problem in preparing and formulating urokinase on an industrial scale or in actually using the same for clinical purposes.
Human albumin has been employed as an additive to urokinas~
so as to improve its stability. However, this can be by no means a break-through solution to the problem just dis-cussed because pure albumin, i.e. a globulin fraction, is difficult to obtain without immunogenic contamination;
pure albumin is expensive; albumin and urokinase form a complex of a high molecular weight under virus deacti-~ Z~7 7~

vating conditions in which urokinase is subjected to heattreatment at 60C for 10 hours together with albumin added to stabilize urokinase; and such stabilizer if added may be effective to a certain extent for protecting urokinase from losing its enzymatic activity upon the lyophilization but cannotPreVentits loss of activity upon actual clinical use.
The physiological activity of urokinase when admi-nistered intravenously to living bodies is promptly retarded by protease inhibitors present in blood (~2-macroglobulin, and a2-plasmin inhibitors and the like), and the metabolic rate of urokinase per se is ve~y hig;~, -es~l.-ing in extremely shortened half-life which does not exceed even several minutes. Nothing has been heretofore propos-d to solve the problem of s~ort half-life of urokinase in blood. - -_ The present inventors have carried out extensive research with a view toward developing derivatives of human-oriyinated non-immunogenic plasminogen activators which will overcome the above-noted drawbac~s of the prior art techniques. As a result, they have succeeded in ind-ing plasminogen activator derivatives whicn are stable and hardly retarded by inhibitors present in blood and hence achi2ve prolonged half-life in blood, thereby leading to the present discovery.
::' SUMMARY OF T~E INVENTION
-An object of the present invention is to provide a derivative of a human-originated non-immunogenic ~ .

7~

plasminogen activator which is stable and exhibits prolong-ed fibrinolytic activity when administered to living bodies.
Another object of the invention is to provide a process for preparing the novel plasminogen activator derivative.
A further object of the invention is to provide a therapeutically acceptable thrombolytic agent comprising the novel plasminogen activator derivative.
These and other objects and advantages of the inven-tion can be attained by the provision of a derivative of a human-originated ncn-immunogeniC plasminogen activator, comprising at least one polyalkylene glycol attached with at least one coupling agent to amino ~cid side chains of the plasminogen activator, the polyalkylene glycol having a molecular weight in the range of 200 -20,000 and option-ally containing one or more alkyl,alkoxy and/or alkanoyl groups as substituents.

l.Z~7~3 -4a-Thus in one aspect the present invention provides a composition for inhibiting the formation of thrombus in a mammalian bloodstream, which composition comprises an effective amount of a nonimmunogenic plasminogen activator comprising at least one polyalkylene glycol moiety chemically bonded with at least one coupling agent to the amino acid side chains of said plasminogen activator, wherein the polyalkylene glycol has a molecular weight in the range of 200-20,000 and is unsubstituted or is substituted with one or more alkyl, alkoxy or alkanoyl groups or a mixture thereof, together with a pharmaceutically acceptable carrier.
BRIEF DESCRIPTION OF THE DR~WINGS
A more complete appreciation of the present inven-tion and many of the attendant advantages thereof will be readily obtained as the invention becomes better understood by reference to the following detailed description when considered in connection with the accompanying drawings, wherein:
FIG. 1 shows the optimum pH ranges of modified high molecular weight urokinase (PEG-DCI-UX) and unmodified high molecular weight urokinase (molecular weight: 54,000) as lZ~7~
_5_ measured in terms of the amidase activity with a synthetic substrate (S-2444), where the open circles correspond to PEG-DCT-UX and the closed circles to unmodified urokinase;
FIG. 2 shows the optimum pH ranges of modified low molecular weight urokinase (PEG-DCT-L-UK) and unmodified low molecular weight urokinase (molecular weight: 33,000) as measured in terms of the amidase activity with S-2444, where the open circles correspond to PEG-DCT-L-UK and the closed circles to unmodified urokinase;
FIG. 3 diagrammatically shows the stability of PET-DCT-UK and unmodified uro~inase at room temperature in either physiological saline or a Ringer solution, where the solid and broken lines indicate the physiological saline and the Ringer solution, respectively, and where the open circles correspond to PEG-DCT-UK and the closed circles to unmodi-fied urokinase; - _ FIG. 4 shows the stability of PEG-DCT-L-UK and un-modified urokinase (molecular weight: 33,000) against freezing, followed by thawing operation repeated 2, 4 and 6 times, where the open circles correspond toPEG-DCT-L-VK
and the closed circles to unmodified urokinase;
FIG. S shows the varied amounts, as a function of time, of a plasmin inhibitor in a plasma fraction Fl of each of two groups of healthy rabbits, one group adminis-tered with PEG-DCT-UK and the other group with unmodified urokinase (molecular weight: 54,000), where the open cir-cles correspond to the PEG-DCT-UK-administered group and the closed circles to the unmodified urokinase-administered group;

-6- 1~7~
FIG. 6 shows the varied amounts, as a function of time, of plasminogen in a plasma fraction F3 of each of two groups of healthy rabbits, one group administered with PEG-DCT-UK and the other group with unmodified urokinase (mole-cular weight: 54,000), where the open circles correspond to the PEG-DCT-UK-administered group and the closed circles to the unmodified urokinase-administered group; and FIG. 7 shows the varied amounts, as a function of time, of plasminogen in a plasma fraction F3 of each of two groups of healthy rabbits, which plasma was treated w1th an acid and then with a base to inactivate a plasmin inhibitor, one group administered with PEG-DCT-UK and the other grou~
with unmodified urokinase (molecular weight: 54,000), where the open circles correspond to the PEG-~CT-UK-administered group and the closed circles to the unmodified urokinase-administered group.
Figure 8 illustrates the substrate specificity of PEG-Modified UK's. _ _ Figure 9a illustrates the stability of modified Urokinase to UK-Inhibi~ors in Human Plasma.
Figure 9b illustrates the effect of Placental UK-Inhibitor on the Urokinase Activity.
Figure 10 illustrates UK activity in Plasma after UX
injection.
Figure 11 illustrates the relative radioactivity in Blood (%1 for 125I-M-Md and 125I-Native-UK.

:

J ~'~

~7- lZ~77~
Figure 12a illustrates a comparison of the immunogenicity of M-Md with that of native UK by Schultz-Dale test.
Figure 12b illustrates a comparison of the immunogenicity o~ M-Md with that of native UK by Passive Cutaneous Anaphylaxis (PCA) reaction.
Figure 13a illustrates a compaxison of the Plasmin Inhibitor in Fl from Citrated Dog Plasma at certain time intervals after injection of Native UK or PEG-UK into a dog.
Figure 13b illustrates a comparison of the Plasminogen in F3 from Citrated Dog Plasma at certain time interYals after injection of Native UK or PEG-UK into a dog.
Figure 13c illustrates a compari50n of the FDP in Dog Serum at certain time intervals after i.v. Injection of Urokinase or PEG-UK in~o a dog.
Figure 14 illustrates a comparison of FDP levels in dogs bearing artificial thrombus in A. femoralis. Dog A had native UK injected into a proximal branch of the thrombus, while Dog B had PEG-UK injected into the same.

- DESCRIPTION OF THE PREFERRED E~ODIMENTS
By the term "human-originated non-immunogenic plas-minogen activator" as used herein are encompassed not on;y urokinase but also tissue plasminogen activators obtained from human tissues such as uterine, tumor and the like.
These human tissue plasminogen activators also contain those obtained by tissue culture or genetic engineerins.
It should be noted that no limitations are imposed on the molecular weishts of these activators so long as they are obtained in the above-described manner. For example, as urokinase which is a plasminogen activator origina.ed from human urine, high molecular weight urokinase (molecular .

7~3 weight: S4,000) and low molecular weight urokinase (molecu-lar weight: 33,000) may be used solely or in combination.
Suitable polyalkylene glycols which may be used in the invention include a polyethylene glycol and a polypro-pylene glycol. In the case of the polypropylene glycol, both straight-chain polypropylene glycols such as those represented by ~O~CH(CH3)CH2O]nH and branched-chain poly-propylene glycols such as those represented by CH3CH2C-{CH20~cH2cH(cH3)o~nH}3 or HIOCH(CH3)CH2~nOCH{cH2~ocH2cH-(C-~3)]nOH}2.
The molecular weights of the polyalkylene glycols may range from 200 to 20,000. Particularly preferred molecular weights are in the range of 500 - 10,000.
The polyalkylene glycols each may o?tionally contain one or more alkyl, alkoxy and/or alkanoyl groups as substituent groups. Typical example,s of the alkyl groups are methyl, ethyl, propyl,stearyl and the like. Typical examples of the alkanoyl groups are acetyl, propibnyl, stearoyl and the like. As a preferred polyalkylene glycol, an unsubsti-tuted or methyl-substituted polyalkylene glycol is useful.
However, of particular interest is the use of methoxy-polyethylene glycol as the polyalkylene glycol. Chemical modification of urokinase(UK) with activated methoxypoly-ethylene glycol(PEG) of MW 5,000 increased the stability of the urokinase and imparted a markedly extended circulation life in rabbits and rats. Also of interest is the surprising fact that PEG-UK has a superior thrombolytic ability as compared to that ~`;

:~Z~77~
g .
of native UK. It appears likely that the superiority of PEG-UK to native UK with respect to the fibrinolytic activation is from the PEG chains which protect the UK
molecule from interation with inhibitors, thus extending its circulation life.

Suitable coupling agents which may be used in the invention and are adapted to attach a polyalkylene glycol to a non-immunogenic plasminogen activator, for example, urokinase, include those capable of reacting with amino acid side chains of the protein to be modifiedand forming chemical bonds therebetween, for example,.acyl azide, cyanuric halides, p-diazoniumbenzyl ether, 3-(p-diazonium-phenoxy)-2-hydroxypropyl ether, dihalogenosuccinic anhydride and the like. The following partial formulae may be given as examples of the coupling structures between a polyalkylene glycol and urokinase through these coupling agents.

-~ -UK

X
_o- ~ -UK

OH
-O-CH2- ~ N2-UK

-o-cH2cH(OH)cH2 ~ -r~2-UK

wherein X is a halogen atom, and UK is a residual part of the urokinase ~olecule.
The novel urokinase derivative according to the invention can ~e prepared by reacting a coupled product of at least one corresponding polyalkylene glycol and at least one coupling agent with urokinase, the polyalkylene glycol having a molecular weisht in the range of 2Q0 -20,000 and optionally containing one or more alkyl , alkoxy and~or alkanoyl groups as substituents.
Typical examples of the polyalkylene ~lycol-cou~lins agent coupled product include oolyalkylene glycol-~,6-.~2~

dichloro-1,3,5-triazine, polyalkylene glycol-4,6-difluoro-1,3,5-triazine, polyalkylene glycol-4-chloro-6-hydroxy~
1,3,5-triazine, polyalkylene glycol-~-(bro~ocarbonyl)-monopropionate, polyalkylene glycol-azidocarbonyl methyl etller, polyalkylene glycol-(p-diazoniumbenzyl) ether, polyalkylene glycol-3-(p-diazoniumphenoxy)-2-hydroxypropyl ether and the like.
When reacting the polyalkylene glycol-coupling agent coupled product with uroki~ase, it is necessary to choose such reaction conditions that the enzymatic acti-vity is held to a minimum loss. Namely, it is desirable to carry out the reaction at low temperatures, for example, at a temperature of 0C to room temperature in an a~ueous solution such as a buffer. Preferred reaction time may range from several minutes to 5 hours. The pH of the buffer is preferably within such a range that the enzymatic activity of urokinase is not lowered, namely, 2 - 10, preferably 5 - 9. However, the preferred pH range may vary depending on the reactivity of each coupling agent employed and/or the nature of an amino acid residue. The modification degreesof amino acid side chains of the plasminogen activator can be controlled by changing the concentration of the polyalkylene glycol activated witn the coupling agent in a reaction medium.
By wày of illustration, monomethyl ether polyethylene glycol-4,6-dicnloro-1,3,5-triazine having an average molecular weight of 5,000 was reacted at pH 7.0 with hig~

lZ~7~

molecular weight urokinase to obtain novel urokinase derivatives, while changing the concentration of the former reactant to 0.4, 4.0 and 6.0 mM, respectively. Unmodified -amino groups of lysine of the resultant urokinase deri-vatives were quantitatively determined using sodium ~,4,6-trinitrobenzenesulfonate. Their modification degrees were investigated on the basis of the results obtained by the quantitative analyses. The modification percentages of the ~-amino groups of lysine, which were reactive with sodium 2,4,6-trinitrobenzenesulfonate, were 6-7% at 0.4~M, about 40% at 4.0 mM and about 60~ at 6.0 mM. In addition, the molecular weights of these reaction products were determined by SDS polyacryl amide gel electrophoresis.
The average molecular weights of the reaction products were about 60,000 at 0.4 mM and about 120,000 at 4.0 m~1. This finding is substantially in conformity with the results obtained above by the quantitative analyses of the E-amino groups of lyslne. Accordingly, it is necessary to conduct the reaction between the polyalkylene glycol activated with the coupling agent and the plasminogen activator at a high pH level and at a high concentration of tne former reagent when increased modification degrees of the plasminogen activator are desired. On the other hand, where decreased modification degrees are preferable, tne xeaction should be effected at a relatively low pH level and at a low con-centration of the coupled product. The modification degrees .. . .

i~`77 ~

may of course be changed by controlling the reaction time.
By suitably combining these reaction conditions, it is possible to obtain the intended novel plasminogen activator derivatives which are stable and have prolongedfibrinolytic activities.
The polyalkylene glycol activated with the coupling agent can be obtained in the following manner. A terminal-substituted polyalkylene glycol-4,6-dihalogeno-1,3,5-triazine is obtained by reacting its corresponding mono-substituted polyalkylene glycol with a cyanuric halide in an anhydrous solvent and in the presenc,e of a base.
A polyalkylene glycol-4-halogeno-6-hydroxy-1,3,5-triazine is formed by first reacting its corresponding polyalkyler,e glycol with cyanuric halide and then treating the resultant reaction product with water. A polyalkylene glycol-acetoazide is formed by reacting an-ani~n of its corre-sponding polyalkylene glycol with ethyl chloroacetate, followed by treating the reaction product with hydrazine, and finally activating the resulting hydrazide with nitrous acid. A polyalkylene glycol-p-diazoniumbenzyl ether or a polyalkylene glycol-3-(p-diazoniumphenoxy)-2-hydroxypropyl ether is obtained by reacting its corresponding polyalky-lene glycol with p-nitrobenzyl chloride or p-nitrophenyl glyceryl ether and, after reducing the nitro group into an amino group, diazotizing the resultant product with nitrous,acid.
After completion of the reaction of the . . .

lZ177~

polyalkylene glycol activated with the coupling agent and the plasminogen activator, the isolation and purification of the reaction product may be effected in a biochemical manner known per se in the art, for example, by using singly or in combination gel filtration, dialysis, ion-exchange chromatography, affinity chromatography and the - like. Preferably, the conjugate is kept as a solution containing a buffer or a physiological salt, by freezins the solution below -20C, or by lyophilizing the solution.
The optimum pH levels for the novel urokinase derivatives prepared above vary depending on the molecular weights and types of polyalkylene glycols emplo~ed, the types of coupling agents employed, the modification degrees of urokinase, the modification conditions and the measure-ment conditions such as substrates used. ~ihen measured in terms of the amidase activity using a synthetic substrate S-2444, the optimum pH level is within the range of about 8 to 9. In the case of nigh molecular weight urokinase modified by reaction with monomethyl ether polyethylene glycol-4,6-dichloro-1,3,5-triazine having an average molecular weight of 5,000 at pH 7.0 and at a temperature of 0C for 3 hou-s, while using the latter reactant at a concentration of 4.0 mM (hereinafter abbreviated as PEG-DC~-UK), the optimum pH as measured in terms of the amidase activity is 8.2 as shown in FIG. 1. The optimum pH of low molecular weight urokinase modified under the same conditions (hereinarter abbreviated as PEG-DCT-L-UK) is, as measured in terms of `

lZ;~ 7~

the amidase activity, 8~2 as shown in FIG. 2.
The novel urokinase derivatives according to the invention have extremely enhanced stability compared with unmodified urokinase. For instance, FIG. 3 shows the results of the residual urokinase activity obtained relative to the passage of time when urokinase was dilu~ed with a Ringer solution or a physiological solution to sucn a concentration as used by dripping administration for clinical application and then allowed to stand at room temperature. As a result, it has been found that PEG-DCT-UK dissolved at a concentration of 105.3 iu/ml in the Rinser solution maintains an activity of 73.4~ at the initiation of the experiment even after 6 hours. In addition, PEG-DCT~UK retains an initial activity of 79.4~ in the physio-logical saline. On the other hand, unmodified urokinase dissolved at a concentration of 76.I iu~l in a Ringer solution keeps only 33.3% in its initial activity even after 2 hours. In a physiological saline, the residual activity is as low as 26.3%.
FIG. 4 illustrates the results obtained by comparing the stability of PEG-DCT-L-UK and unmodified low moiecula~
weight urokinase, both in the course of freezing, followed by thawing operation. The results confirm th~t th2 supe-b stability of PEG-DCT-L-UK is attained even under such conditions.
Furthermore, the novel urokinase derivatives according to the invention have prolonged urokinase ac~i-lZ:~`7~3 vities in blood as compared with unmodified urokinase.For example, the effectiveness of urokinase was investigated by administering 8,000 iu/kg of each of PEG-DCT-UK and unmodified urokinase to healthy rabbits by intravenous injection, sampling blood periodically before the dosage and after the lapse of 4 hours, and measurins the amounts of the plasmin inhibitor and plasminogen present in the citrated plasma. The results are shown in FIGS. 5, 6 and 7. From these results, it has been con-firmed that PEG-DCT-UK alters to a substantial extent biochemical parameters such as a plasmin inhibitor and plasminogen and maintains the parameters at lowered values for an extended period of time as compared with unmodified urokinase.
Therefore, the ~;~ urokinase derivatives accorâing to the invention are excellent plasminog~n activators which, while retaining plasminogen-activating potency as unmodified urokinase does, have surmounted the shortcomings of urokinase such as poor stability and short half-life in blood.
The urokinase derivatives according to the invention may be used as pharmaceutical products for the treatment of a variety of diseases stenlmed from hyper-coagulability of blood such as arterial and venous throm-boses, coronary artery clotting, myocardial infarction, intracerebral infarction, pulmonary embolism, nephritis and the like. These urokinase derivatives can be suitAbly . . .

L77~

administered by intravenous injection or dripping or by an oral route. The intravenous injection is particulary preferred. As dosable forms, preferably usable is a lyophilized form. When supplied eithex in a neat state or with a physiological salt such as a salt of a Ringer solution or sodium chloride, the lyophilized products may be used for clinical application by dissolution with sterile distilled water or further dilution with sterile distilled water in order to adjust their osmotic pressure, prior to the actual use. Since the novel urokinase de-i-vatives according to the invention are stable,they do not require a stabilizer such as albumin. However, the addi-tion of such a stabilizer does not cause any problem or inconvenience. An excipient may also be added in subjec~-ing the novel urokinase derivatives to lyophilization.
The above description genera-lly ~escribes the present invention. A more complete understanding can be obtained by reference to the following examples which are provided for purposes of illustration only and are not intended to be limiting. The modification procedure may be carried out in any one of the preparation steps of plasminogen activators.
Example 1 Monomethyl ether polyethylene glycol-4,6-dicnloro-1,3,5-triazine Polyethylene glycol monomethyl ether having an average molecular weight of 5,000 (25.0 g; O.OS mole) was ~ 77:~

dissolved with warming in dry benzene (200 ml). After cooling the resultant solution to room temperature, anhydrous sodium carbonate (5.0 g) and cyanuric chloride (2.75 g; 0.015 mole) were added, and the mixture was stirred overnight at room temperature. After com?letion of the reaction, the reaction mixture was filtered, and petroleum ether t600 m1) was added to the filtrate. The resulting precipitate was collected by suction filtration and washed with a small amount of petroleum ether. The precipitate was purified by being reprecipitated three times from dry benzene and petroleum ether to remove excess cyanuric chloride, thereby stoichiometrically obtaining monomethyl ether polyethylene glycol-4,6-dichloro-1,3,5-triazine as white powder. The product after hydrolyzed showed qualitative reaction characteristics of chlorine ions (AgNO3).
- Similarly, polyethylene glycol monomethyl ethers having average molecular weights of 550, 700, 2,000 and 20,000, respectively, were each reacted with cyanuric chloride to stoichiometrically obtain monomethyl ether polyethylene glycol-4,6-dichloro-1,3,5-triazines having the corresponding average molecular weights.
Example 2 Monomethyl ether polyethylene glycol-4,6-dichloro-1,3,5-triazines (average molecular weights o~ their polyethylene glycol moieties: 10,000 and 15,000) Polyethylene glycol monomethyl ether having an ~77~

average molecular weight of 10,000 (25.0 g; 0.025 mole) was taken up with warming in dry benzene (200 ml~. After cooling the resultant solution, anhydrous sodium carbonate (2.5 g) and cyanuric chloride (1.38g; 0.0075 mole) were added. The resultant mixture was stirred overnight at 33C. After completion of the reaction, undissolved matter was removed by filtration. The filtrate was added with n-hexane (about 600 ml) to induce reprecipitation. Dry benzene (100 ml) was added to the precipitate which was then warmed and dissolved. n-~exane (500 rll) was then added to induce reprecipitation. This procedure was repeated three times.- The thus obtained precipitate was dried overnight at 50C in vacuo, thereby stoichiometri-cally obtaining monomethyl ether polyethylene glycol-4,6-dichloro-1,3,5-triazine (the average molecular weight of the polyethylene glycol moiety: lO,~OOO)~as while powder.
Polyethylene glycol monomethyl ether having an average molecular weight of 15,000 was similarly reacted with cyanuric chloride to stoichiometrically obtain monomethyl ether polyethylene glycol-4,6-dichloro-1,3,5-triazine (the average molecular weight of the polyethylene glycol moiety:
15,000).
Example 3 Monostearyl ether polyethylene glycol-4,6-dichloro-1,3,5-triazine Polyethylene glycol monostearyl ether having an average molecular weight of 3,200 (lo .0 g; 0.005 mole) was 77~

dissolved in dry benzene (200 ml), followed by the addi-tion of anhydrous sodium carbonate (5.0 g) and cyanuri~
chloride (2.75 g; 0.015 mole) under stirring. The resul-tant mixture was stirred overnight at room temperature.
After completion of the reaction, undissolved matter was removed by filtration. The filtrate was added with n-hexane (about 600 ml) to induce reprecipitation. Dry benzene (100 ml) was added to the precipitate to dissolve the latter. n-Hexane (500 ml) was added to induce reprecipitation. This procedure was repeated three times, thereby stoichiometrically obtaining monostearly ether polyethylene glycol-4,6-dichloro-1,3,5-triazine (the average molecular weight of the polyethylene glycol moiety:
3,200) as white powder.
Example 4 Polyethylene glycol-4-chloro-6-hydroxy-1,3,5-triazine Polyethylene glycol having an average molecular weight of 6,000 (33.6 g) was dissolved with warming in dry benzene (150 ml). After cooling the resultant solution, anhydrous sodium carbonate (1.6 g) and cyanuric chloride (0.74 g) were added, and the resultant mixture was stirred overnight at room temperature. Thereafter, water (1.0 ml) was added, followed by stirring the mi~ture at room tem-perature for 6 hours and then at 40C for an overnight period. Undissolved matter was removed by centrifuge (2,000 ppm; 10 minutes), and the supernatant was subjected 7 7~

to condensation under reduced pressure. The residue was taken up with warming in dry benzene, and the soIvent w~s then evaporated. This procedure was further repeated twice. The residue was dried under reduced pressure, thereby obtaining polyethylene glycol-4-chloro-6-hydroxy-1,3,5-triazine (the average molecular weight of the polyethylene glycol moiety: 6,000).
Polyethylene glycols having different average molecular weights of 1,000 and 4,000 were each reacted in the same manner as above with cyanuric chloride and water, resulting in the stoichiometric formation of polyethylene glycol-4-chloro-6-hydroxy-1,3,5-triazines having their correspondins average molecular weishts.
Example 5 Stearoylpolyethylene glycol-4,6-dichloro-1,3,5-triazine - Polyethylene glycol monostearate having an average molecular weight of 2,700 (6.765 g) was dissolved in anhydrous benzene (100 ml), followed by the addition of anhydrous sodium carbonate (2.5 g). While stirring the resultant mixture, cyanuric chloride (1.38 g~ was further added. The resulting mixture was stirred overnight at room temperature and then filtered. The filtrate was concentrated under reduced pressure, and the residue was dried under reduced pressure, thereby stoichiometrically obtaining stearoylpolyethylene glycol-4,6-dlchloro-1,3,5-~riazine (the average molecular weight of the polyethylere 77~

glycol moiety: 2,700) as a white waxy substance.
Example 6 Polypropylene glycol-4-chloro-6-hydroxy-1,3,5-triazine Polypropylene glycol having an average molecular weight of 1,000 (4.0 g) was taken up in anhydrous benzene (50 ml), followed ~y the addition of anhydrous sodium carbonate ~1.27 g). Cyanuxic chloride (0.552 g) was further added with stirring. After stirring the resultant mixture overnight at room temperature, water (1 ml) was added. The mixture was stirred at room temperature for further 6 hours. The reaction mixture was filtered, and the filtrate was concentrated under reduced pressure.
The residue was added with anhydrous benzene and anhydrous sodium sulfate. The mixture was thereafter stirred at room temperature for 10 minutes. ~ter~~iltration of the mixture, the solvent was evaporated from the filtrate.
The residue was dried under reduced pressure, thereby stoichiometrically obtaining polypropylene glycol-4-chloro-6-hydroxy-1,3,5-triazine (the average molecular weight of the polypropylene glycol moiety: 1,000) as colorless viscous oil.
Following the same procedure as described above, there were obtained a polypropylene glycol-4-chloro-6-hydroxy-1,3,5-triazine (the average molecular weight of the polypropylene glycol moiety: 4,000) and anothe~ poly-propylene glycol-4-chloro-6-hydroxy-1,3,5-triazine (the ~z~

average molecular weight of the polypropylene glycol moiety: 10,000).
The polypropylene glycols employed in the above examples were of a straight-chain type, namely, those respresented by the formula HO[CH(C~3)CH2O]nH.
Example 7 Polypropylene glycol-4-chloro-6-hydroxy-1,3,5-triazine Following the same procedure as in Example 1, poly-propylene glycol-4-chloro-6-hydroxy-1,3,5-triazine (the average molecular weight of the polypropylene glycol moiety: 4,000) was stoichiometrically obtained as a white substance from polypropylene glycol having an average molecular weihgt of A,000 ~8.0 g), anhydrous benzene t80 ml), anhydrous sodium carbonate (0.636 g) and cyanuric chloride (0.368 g).
. The polypropylene glycol used in this example was of a branched-chain type, namely, those re~resented by the formula CH3CH2C{CH2O[CH2CH(CH3)O]nH}3-Furthermore, using polypropylene glycol representedby the formula, CH2O[CH2cH(cH3)O]nH
CHO[CH2CH(CH3)O]nH
CH2O[CH2CH(cH3)O~nH
and naving an average molecular weight of 3,000, there was obtained polypropylene glycol-4-chloro-6-hydroxy-1,3 "-triazine (the average molecular weight of the polypropylene ~2~7~

glycol moiety: 3,000).
Example 8 ~lonomethyl ether polyethylene glycol methoxycarbohydrazide Polyethylene glycol monomethyl ether having an average molecular weight of 5,000 (13.3 g; 0.0027 mole) was taken up in anhydrous tetrahydrofuran (400 ml) under nitrogen. A small amount of diphenylacetic acid was added as an indicator, and n-butyl lithium was dropped under ice-cooling until the reaction solution turned to pale yellow.
Thereafter, ethyl chloroacetate (5 ml; 0.0~7 mole) was dropped at room temperature, and the resultant solution was stirred overnight and then refluxed for one hour.
After completion of the reaction, the solvent was driven off under reduced pressure,and the residue was taken up in aqueous acetone(200 ml). The resul~nt solution was treated with charcoal. After filtration and subsequent concentration of the filtrate, benzene was added. After removing water by azeotropic distillation, the residue was reprecipitated from benzene and n-hexane to obtain yellowish powder. The thus obtained powder was dissolved in methanol (150 ml) and tnen added with hydrazine hydrate (15 ml?. The mixture was heated overnight under reflux conditions. After driving off the solvent under reduced pressure, water (150 ml) was added, and excess hydrazine was removed by azeotropic distillation. Water, still remaining in the residue, was removed azeotropically 7~

together with benzene. The residue was again dissolved in benzene and dried with anhydrous sodium sulfa~e.
Thereafter, the solvent was driven off, and the residue was dissolved in warm benzene. The benzene solution was treated with charcoal and then concentrated. The reaction product was reprecipitated twice from benzene and n-hexane, treated with charcoal again and with silica gel and then reprecipitated from benzene and n-hexane, thereby obtain-ing monomethyl ether polyethylene glycol methoxycarbo-hydrazide as white powder. In the same manner, a variety of hydrazides were obtained using polyethylene glycol monomethyl ethers havlng average molecular weights of 550, 700, 2,000, 10,000, 15,000 and 20,000, respectively, as starting materials.
Example 9 Monomethyl ether polyethylene glYCo1-4, 6-dichloro-1,3,5-triazine-modified urokinase (PEG-DCT-UK) A 0.1 M phosphate buffer of pH 7.0 (2.0 ml) W2S
added under ice-cooling to 0. 61 ml of a urokinase solution (molecular weight: 54,000; 66,300 iu/ml). Thereafter, monomethyl ether polyethylene glycol-4,6-dichloro-1,3,5-triazine (the average molecular weight of the polyethylene glycol moiety: 5,000) was further added in such an amount as to bring the concentration of the triazine to 4 mM.
The mixture was reacted under ice-cooling for 3 hours.
After completion of the reaction; the reaction solution i;~177~

was transferred into a dialyzing tube, and excess triazine derivative was removed by dialysis. The dialysis was carrled out under ice-cooling for 3 hours against a 0.1 M
phosphate buffer (pH 7.2) and then for further one hour against physiological saline. The content in the tube ~7as then filled up to 4 ml and stored in a frozen state at -80C. The urokinase activity of the thus obtained mono-methyl ether polyethylene glycol-4,6-dichloro-1,3,5-triazine-modified urokinase (PEG-DCT-U~) was determined to be 4,550 iu/ml by the fibrin plate method. Therefore, the total activity was 13,200 iu. Since the activity of the starting urokinase was 40,443 iu, an activity drop of 55% was recognized.
The above procedure was repeated except that the monomethyl ether polyethylene glycol-4,6-dichloro-1,3,5-triazine (the average molecular weight~~of the polyethylene glycol moiety: 5,000) was replaced by monomethyl ether polyethylene glycol-4,6-dichloro-1,3,5-triazines (the average molecular weights of the polyethylene glycol moieties: 550, 700 and 2,000, respectively), thereby obtaining modified urokinases having urokinase activities ....
Of 9,800, 10JOOO and 6,500 iu/ml as determined by the fi~rin plate method.
Example 10 Monomethyl ether polyethylene ~lycol-4,6-dichloro-1,3,5-triazine-modified low molecular ~ .
weight urokinase (PEG-DCT-L-U~) A 0.1 M phosphate buffer of pH 7.0 (4.7 ml) was ~ .
.

1~77~

added under ice-cooling to 0.2 ml of a low molecular weight urokinase solution (molecular weight: 33,000; 567,828 iu/
ml). Thereafter, monomethyl ether polyethylene glycol-4,6-dichloro-1,3,5-triazine (the average molecular weight of the polyethylene glycol moiety: 5,000) was further added in such an amount as to bring the concentration of the triazine derivative to 4 mM. The mixture was reacted under ice-cooling for 3 hours. After completion of the reaction, the reaction solution was transferred into a dialyzing tube, and excess triazine derivative was removec by dialysis. The dialysis was carried out under ice-cooling for 3 hours against a 0.1 M phosphate buffer (pH
7.2) and then for further one hoùr against physiological saline. After the dialysis, the content was filled u? to 8 ml and stored in a frozen state at -80C. The urokinase activity of the thus obtained monomethyl ether polyethylene glycol-4,6-dichloro-1,3,5-triazine-modified low molecular weight urokinase (PEG-DCT-L-UK) was determined to be 10,300 iu/ml by the fibrin plate method. Thus, the total activity was 82,400 iu. Since the activity of the starting urokinase was 113,566 iu, an activity drop of 27.4~ was recognized.
Example 11 Monomethyl ether polyeth~lene ~lycol-4,6-dichloro-1,3,5-triazine-modified urokinase By repeating the procedure of Example 9 except t~at the concentration of each monomethyl ether polyethvlene .

~77~

glycol-4,6-dichloro-1,3,5-triazi~e was changed to 0.4 mM, there were obtained, with modification degrees different from that achieved in Example 9, monomethyl ether poly-ethylene glycol-4,6-dichloro-1,3,5-triazine-modified urokinases (the average molecular weights of the poly-ethylene glycol moieties: 550, 700, 2,000 and 5,000, respectively). Their urokinase activities were 8,670, 8,900, 6,770 and 8,670 iu/ml as measured by the fibrin plate method.
Example 12 Monomethyl ether polyethylene carbomethylazide-modified urokinase (1) To 200 mg of the monomethyl ether polyethylene glycol methoxycarbohydrazide prepared in Example 8 (the average molecular weight of the polyethylene glycol moiety:
5,000) were added under ice-coolin~ 1 ~_hydrochloric acid (2 ml) and then a 0.008 N aqueous solution of sodium nitrite (1 ml). The resultant mixture was stirred at room temperature for 20 minutes, and a 1 N aqueous solution of sodium hydroxide (2 ml) was further added to neutralize the mixture. The resultant solution of monomethyl ether polyethylene glycol carboxymethylazide was stored at 0C.
(2) A 0.1 M phosphate buffer of pH 8.0 (3.656 ml) was added to 1 ml of a urokinase solution (molecular weight: 33,000; 45,600 iu/ml). Thereafter, 0.435 ml of the monomethyl ether polyethylene glycol carbomethylazide solution prepared in the procedure (1) above was added .. .. .

~Z~7~3 -under mild stirring. The mixture was reacted at room temperature for 2 hours. The resulting reaction solution was then transferred into a dialyzing tube and dialyzed under ice-cooling for 4 hours against a 0.1 M phosphate buffer (pH 7.2). The content was filled up to 8 ml. The resultant solution was stored in a frozen state at -80C.
The urokinase activity of the thus obtained monomethyl ether polyethylene glycol carbomethylazide-modified uroki-nase was determined to be 7,300 iu/ml by the fibrin plate method. Compared with unmodified urokinase, an activity increase of 28% was recognized.
E~ample 13 Monomethyl ether polyethylene glycol-4,6-dichloro-1,3,5-triazine-modified urokinase To 0.61 ml of a urokinase solution (molecular weight: 54,000; 101,167 iu/ml) were added under ice-cool`ng -a 0.1 M phosphate buffer of pH 7.0 (2.0 ml) and then under mild stirring monomethyl ether polyethylene glycol-4,6-dichloro-1,3,5-triazine (the average molecular weight of the polyethylene glycol moiety: 10,000). The triazine derivative was added in such an amount as to bring the concentration to 0.1 mM. The mixture was reacted for 3 hours under ice-cooling. After completion of the reaction, the reaction solution was transferred into a dialyzing tube, and excess triazine derivative was removed by dialy-sis. The dialysis was carried out under ice-cooling for 3 hours against a 0.1 M phosphoric acid bùffer added wi-n . . .

~Z~'77~3 .

0.035 v/v % of ethyl amine (pH8.0) and then for further 2 nours against a O.lM phosphate buffer (pH 7.2). The con-tent was adde~ with 3 w/v % bovine serum albumin (0.1 ml) and then filled up to 4.0ml with a 0.1 M phosphate buffer (pH 7.0~. The resultant solution was stored in a frozen state at -80C. The activity of the thus obtained mono-methyl ether polyethylene glycol-4,6-dichloro-1,3,5-triazine-modified urokinase was determined to be 1~,0~8 iu/ml by the fibrin plate method. Therefore, the total activity was 40,11~ iu, and the activity drop was 35%. Similar modi-fied urokinases were obtained by changing the concentration of the monometnyl ether polyethylene glycol-4,6-dichloro-1,3,5-triazine (the average molecular weight of the poly-ethylene glycol moiety: lO,OOO)to 0.4and 1.0 ~1, respectively.
Their urokinase activities were 8,7~4 and 6,634 iu/ml.
Furthermore, monomethyl ethe~ por~ethylene glycol-
4,6-dichloro~1,3,5-triazine (the average molecular weight of the polyethylene glycol moiety: 15,000~ and urokinase were similarly reacted, thereby obtaining monomethyl ether polyethylene glycol-4,6-dLchloro-1,3,5-triazine-modified urokinàse (the average molecular weight of the polyethylene glycol moiety: 15,000). The activities of the modified urokinases obtained by changing the concentration of the triazine derivative to 0.1, 0.4 and 1.0 ~I, respectively, were 11,388, ~,580 and 7,176 iu/ml.

'771~

Example 14 Monostearyl ether polyethylene glycol-4,6-dichloro-1,3,5-triazine-modified urokinase To 0.2 ml of a urokinase solution (molecular weight:
54,000; 101,167 iu/ml) were added under ice-cooling a 0.1 M
phosphate buffer of pH 7.0 (0.66 ml) and then monostearyl ether polyethylene glycol-4,6-dichloro-1,3,5-txiazine (the average molecular weight of the polyethylene glycol moiety:
3,200). The triazine derivative was added in such an amount as to bring the concentration to 2 mM. The mixture was reacted under ice-cooling for 3 hours. After comple-tion of the reaction,~the reaction solution was transferred into a dializing tube and subjected to dialysis to remove excess triazine derivative. The dialysis was carried ou~
under ice-cooling for 4 hours against a 0.1 M phosphate buffer of pH 7.2. The content was ~dde~ with a 3.0% aque-ous solution of bovine serum albumin (0.1 ml) and then filled up to 4.0 m~ with a 0.1 M phosphate buffer of pH
7Ø The resultant solution was stored in a frozen state at -80C. The urokinase activity of the thus prepared monostearyl ether polyethylene glycol-4,6-dichloro-1,3,5-triazine-modified urokinase (the average molecular weight of the polyethylene glycol moiety: 3,200) was determined to be 2,826 iu/ml by the fibrin plate method. Since the total activity was 11,304 iu, the activity of the thus modified urokinas~e was 55.9% of that of t~e starting urokinase. The activities of tle modified urokinases 7 ~

obtained by changing tne concentration of the monostearyl ether polyethylene glycol-4,6-dichloro-1,3,5-triazine to 4, 6 and 8 mM, respectively, were 2,351, 1,430 and 1,059 iu/ml.
Example 15 .
Polyethylene glycol-4-chloro-6-hydroxy-1,3,5-triazine-modified urokinase To 1 ml of a urokinase solution (molecular weight:
54,000; 45,600 iu/ml) were added under ice-cooling a 0.05 M
phosphate buffer of pH 9.2 (4.0 ml) and then polyethylene glycol-4-chloro-6-hydroxy-1,3,5-triazine (2.34 mg). The average molecular weight of the polyethylene glycol moiety of the triazine derivative was 6,000. The mixture was reacted under ice-cooling for 3 hours. After completion of the reaction, the reaction solution was transferred into a dialyzin~ tube, and excess txiaz~ne derivative was removed by dialysis. The dialysis was carried out under ice-cooling for one hour against a 0.05 M phosphate buffer of pH 9.2 and then for further 3 hours against a 0.1 M
phosphate buffer of pH 7.2. The content was filled up to 8.0 ml with a 0.1 M phosphate buffer of pH 7.2. The resultant solution was stored in a frozen state at -80C.
The urokinase activity of the thus obtained polyethylene glycol-4-chloro-6-hydroxy-1,3,5-triazine-modified urokinase (the average molecular weight of the polyethylene glycol moiety: 6,000) was determined to be 460 iu/ml by the fibrin plate method.

~'77~'~

Similarly, there were obtained other polyethylene glycol-4-chloro-6-hydroxy-1,3,5-triazine-modified urokinases (the average molecular weights of the polyethylene glycol moieties: 4,000 and 1,000, respectively).
Example 16 Stearoyl polyethylene glycol-4,6-dichloro-1,3,5-triazine-modified urokinase .
- To 0.5 ml of a uro~inase solution (molecular weisht:
54,000; 101,167 iu/ml) were added under ice-cooling a 0.1 phosphate buffer of pH 7.0 (1.5 ml) and then 0~05 ml of a dioxane solution of stearoyl polyethylene glycol-4,6-dichloro-1,3,5-triazine (270 mg/ml) (the average molecular weight of the polyethylene glycol moiety: 2,700). The mixture was reacted under ice-cooling for 3 hours. ~fter completion of the reaction, the reactio~ solution was transferred into a dia~yzing ~ube~and-~ùbjected to dialysis to remove excess triazine derivative. The dialysis was carried out under ice-cooling for 4 hours against a 0.1 M
phosphate buffer of pH 7.2. The content was filled up to ~ ml with a pnosphate buffer. The resultant solution was stored in a frozen state at -80C.
The thus obtained stearoyl polyethylene glycol-4,6-dichloro-1,3,5-triazine-modified urokinase was found, a result of the urokinase activity measurement by the fibrin plate method, to have an activity of la6% of that of the starting urokinase.

~Z177~

Other stearoyl polyethylene glycol-4,6-dichloro-1,3,5-triazine-modified urokinases having different modi-fication degrees were obtained by reaction with urokinase using a dioxane solution of stéaroyl polyethylene glycol-4,6-dichloro-1,3,5-triazine (270 mg/ml) in amounts of 0.1 and 0.2 ml, respectively. The average molecular weight of the polyethylene glycol moiety of the triazine derivative was 2,700. The thus prepared modified urokinases had activities of 106 and 101% of that of the starting urokinase.
Example 17 Polypropylene glycol-4-chloro-6-hydroxy-1,3,5-triazine-modified urokinase To 0. 5 ml of a urokinase solution (molecular weight:
54,000; 101,167 iu/ml) were added under ice-cooling a Ool M phosphate buffer of pH 7.0 (I.5 m~) and then 0.05 ml of a dioxane solution of the polypropylene glycol-4-chloro-6-hydroxy-1,3,5-triazine (1-00 mg/ml) obtained in Example 6. The average molecular weight of the polypro-pylene glycol moiety of the triazine derivative was 1,000.
The mixture was reacted under ice-cooling for 3 hours.
Ater completion of the reaction, the reaction solution was transferred into a dialyzingtube and subjected to dialysis to remove excess triazine derivative. The dialy-sis was carried out under ice-cooling for 4 hours against a 0.1 M phosphate buffer of pH 7.2. The content was filled up to 5 ml with a phosphate buffer and then stored lZ~77~

in a frozen state at -80C. The thus obtained polypro-pylene glycol-4-chloro-6-hydroxy-1,3,5-triazine-modified urokinase was found to retain an activity of 96.4~ of that of the starting urokinase by the fibrin plate method.
Other polypropylene glycol-4-chloro-6-hydroxy-1,3,5-triazine-modified urokinases having different modi-fication degrees were similarly obtained by using a dioxane solution of polypropylene glycol-4-chloro-6-hydroxy-1,3,5-triazine (100 mg/ml) in amounts of 0.1 and 0.2 ml, respectively. The average molecular weight of the polypropylene glycol moiety of the triazine deriva-tive was 1,000. The modified urokinases showed activities of 99.3 and 106.8% in comparison with that of the starting unmodified urokinase.
ExamPle 18 ;
Polypropylene glycol-4-cnloro-6-~droxy-- . 1,3,5-triazine-modified urokinase Polypropylene glycol-4-chloro-6-hydroxy-1,3,5-triazine-modified urokinases having different modification degrees were obtained exactly in the same manner as in Example 3 with use of a dioxane solution containing 400 mg/
ml of the polypropylene glycol-4-chloro-6-hydroxy-1,3,5-triazine obtained in Example 7 in amounts of 0.05, 0.1 and 0.2 ml, respectively. The average molecular weight of the polyethylene glycol moiety of the triazine derivative was 4,000. Their urokinase activities were found to be Rg,7, 100.0 and 98.9% of the that of the 7 ~

starting urokinase by the fibrin pla~e method.
Example 19 Optimum pH for modified urokinase as measured in terms of amidase activity PEG-DCT-UK obtained in Example 9 and unmodified urokinase were each diluted with physiological saline containing 0.1% human albumin to obtain two solutions of 167 iu/ml. Each of the solutions were divided into por-tions of 0.3 ml which were added with 50 mM tris-HCl buffers of different pH levels (each 1.0 ml) and tnen with a synthetic substrate S-2444 (pyrGlu-Gly-Arg-p-nitro anilide made by Kabi Corporation). The resultant mi:~ture;
were incubated at 37C for 10 minutes. The reactions were then stopped by the addition of 30% acetic acid. Their absorbance was measured at 405 nm. As a result, the optimum pH levels for PEG-DCT-UK and unmodified urokinase were 8.2 and 8.5, respectively, as measured in terms of the amidase activity when the synthetic substrate S-2444 was employed. The results are shown in FIG. 1.
The optimum pH levels for PEG-DCT-L-UK obtained in Example 4 and unmodified low molecular weight urokinase were determined similarly in terms of the amidase activity and we~e found to be 8.2 and 8.4, respectively. The results are shown in FIG. 2.

~2~7'71~

Example 20 Stabilit of modified urokinase at room Y
temperature PEG-DCT-UK obtained in Example 9 was diluted with either a Ringer solution or physiological saline to a concentration of 105.3 iu/ml. For the sake of comparison, unmodified urokinase was also diluted with either a Ringer solution or physiological saline to a concentration of 76.1 iu/ml. 3.5 ml of each of the thus diluted solutions was allowed to stand at room temperature (27~C) for 6 hours. The residual urokinase activity was periodically measured by the fibrin pla~e method until such time tha~
6 hours elapsed. The extent of activity loss of the modified urokinase according to the invention was by far smaller than that of unmodified urokinase. The results are shown in FIG. 3.
-Example 21 -Stability of modified urokinase against freezing and thawing processing PEG-DCT-L-UK obtained in Example 10 was diluted with physiological saline to a concentration of 2Q0 iu/ml.
For the sake of comparison, unmodified low molecular weight urokinase was diluted with physiological saline to a concentration of 200 iu/ml. Each of the thus diluted solutions was frozen to -80C and subsequently thawed at room temperature. The freezing and thawing operations were repeated 0, 2, 4 and 6 times. The residual urokinase 1;Z~'77;~

activity was measured using a synthetic substrate S-244~
(made by Kabi Corporation). The extent of activity loss of the modified urokinase according to the invention was extremely low as compared with that of unmodified low molecular weight urokinase. The results are shown in FIG. 4.
Example 22 Eight healthy male rabbits (Japanese white) each having a body weight of 2.6 - 3.1 kg were divided into two groups. One group was dosed with 8,000 iu/kg of PEG-DCT-UK and the other with 8,000 iu/kg of unmodified urokinase, both through the ear veins. Blood was collec-ted from the ear veins before the dosage and at the time of 1, 2 and 4 hours after the dosage. Blood samples W21-e added with a sodium citrate solution to separate plasma.
After wash-out of the modified and unmo~ified urokinases for 18 days, the two groups were crossed with each other, and similar experiments were carried out to take plasma samples. A part of each of the plasma samples thus obtained was separated and subjected to affinity column chromatography using Lysine Sepharose 4B, thereby obtain-ing a plasmin inhibitor fraction (fraction Fl) and a plasmin and plasminogen fraction (fraction F3). Chan~es in the amount of the plasmin inhibitor in the fraction Flalong the passage of time were determined by adding plasmin to the fraction Fl and measuring the amount of residual plasmin which was not adversely affected b~ the inhibitor, with a svnthe'ic ,tl 7-.lB

substrate S-2251 (made by Kabi Corporation). The plasmin inhibitor from the PEG-DCT-UK-administered group showed its apparent decrease and slower recovexy. In the unmodi-fied urokinase-administered group, the decrease in the plasmin inhibitor was rather less, and its time course changes were not made clear. The results are shown in FIG. 5.
On the other hand, the amount of plasminogen in the fraction F3 was determined by adding urokinase to the fraction F3 to produce plasmin and measuring the amount of the thus produced plasmin by a synthetic substrate S 2251. In the PEG-DCT-UK-administered group, plasminogen decreased to about 50% after 2 hours and thereafter re-covered gradually. However, in the unmodified urokinase-administered group, plasminogen decreased up to about 65~
after one hour but did not proceed ~to decrease any fur.her.
-There was observed a tendency of plasminogen being recov~
ered even 2 hours after the dosage. The results are illustrated in FIG. 6.
Furthermore, a part of each plasma was sampled ana treated with an acid to pH 5.2 so as to deactivate the inhibitor therein. It was then neutrali~ed, and the a~ount of plasminogen present in the acid treated plasma was measured by adding urokinase to the acid treated plasma to produce plasmin and measuring the amount of the thus produced plasmin using a synthetlc SuDstrate S-2251.
In the PEG-DCT-UK-administered gro~lp, plasminogen decreased to about 70% after 2 hours but showed its gradual recovery thereafter. In the unmodified urokinase-administered group, however, the amount of plasminogen was decreased to about 84% after one hour but was not decreased any further.
Thereafter, the amount of plasminogen was increased gra-dually. The results are also shown in FIG. 6. Incidental-ly, no plasmin activity was detected from the fraction F3.
Example 23 Lyophilized_product suitable for the preparation of injectable formulations The PEG-DCT-UK solution obtained in Example 9 was concentrated by a membrane filter (Amicon ~el: trademark~
and then added with physiological saline and a O.OS M
phosphate buffer. The resultant mixture was filtered aseptically using a membrane filter. The filtrate was poured in portions into sterilized vials and then lyophi-lized. The fibrinolytic activity of the lyophilized product of PEG-DCT-UK suitable for the preparation of injectable formulations was found to be 57,000 iu/vial by the fibrin plate method.
Example 24 L o hilized product suitable for tne preparation . Y P
of injectable formulations The PEG-DCT-UK solution obtained in Example 9 was concentrated by ~ membrane filter (~micon Gel: trademark) and then added with physiological saline. The resultant mixture was filtered aseptically using a membrane filter.

; 7 ~r~

- The filtrate was poured in portions into sterilized vials and then lyophilized. The fibrinolytic activity of ~he thus obtained lyophilized product of PEG-DCT-UK suitable for the preparation of injectable formulations was found to be 67,000 iu/vial by the fibrin plate method.
In the following experiments Urokinase (MW 54,000) was obtained from Japan Chemical Research Co., Ltd. PEG
#5,000 (MN = 4,700, MW/MN = l.08) was obtained from Nishio Industry Co., Ltd. Cyanuric chloride was obtained from Kanto Chemical Co., Inc. Fibrinogen and thrombin fcr fibrin plate was obtained from Sigma Chemical Co. and Mochida Pharmaceutical Co., Ltd.
respectively. S-2251 and S-2444 were obtained from Kabi Diagnostica. TNBS (trinitrobenzene sulfonic acid sodium salt) was obtained from Wako Pure Chemical Industry, Ltd. Lysine-Sepharose 4B and Sephadex G-200 were obtained from Pharmacia Fine Chemicals. Nal25I
was obtained from Amersham. GGA-MCA (L-glutaryl-glycyl-L-argir.ine-4-methylcoumarin-7-amide) was obtained from Protein Research Founda~ion.
Urokinase dissolved in O.lM phosphate buffer (P~), pH 7.0, was reacted with PEG #5,000 activated with cyanuric chloride by the method of A~uchowski et al at * trade mark.

3~2~i7~'~

4 for 4 hr. Concentration of PEG was adjusted to 1.7 mM for L-Md, 2 mM for M-Md and 5 mM for H-Md which represent low, medium and high degrees of modification, respectively. Dialysis against 0.05 m PB, pH 5.0, containing 0.9% ~aCl followed by concentration and gel filtration using Sephadex G-200 afforded modified UR's. After adjusting pH to 7.0, UK'5 were lyophilized in small portions for experimental convenience. Lysine residues in M-Md were determined with TNBS by the method of Habeeb. UK's were assayed either by the standard fibrin plate method or by the synthetic substrate method. Plasma was fractioned into Fl, F2 and F3 with Lysine-Sepharose 4B by the method of Igarashi et al. Native UK and M-Md were labelled with 12SI by the procedure of Hunter and Greenwood.
Immunological properties of native UK and M-Md were compared according to the procedures of Arai et al.

Example 2S - SUBSTRATE SPECIFICITY OF PEG-MODIFIED UK's Activation of PEG #5,000 with cyanuric chloride followed by reaction with native UK caused a covalent attachment of PEG's on the lysine residues of UK
protein. Three types of modified UK's, L-Md, M-Md and H-Md, were prepared by changing PEG concentration during the reaction. Their enzymatic activitie~ depend on the assay method which sugges~s that PEG chains ~Z~ 7~

presen~ steric hindrance to bulky substrat~s such as plasminogen in fibrin plate while not to the low molecular weight ones such as GGA-MCA.
Attention was specifically focused on M-Md and the pH Optimum of M-Md was found to be 8.16 and that of native UK to be 8.51 using S-2444 as a substrate in 0.05M Tris HCI buffer at 37C. The molecular weight (MW) of M-Md was calculated to be about 120,000 daltons from the result of lysine residue determination with TNBS. As PEG is a kind of neutral detergent the apparent MW obtained from SDS-PAGE of gel filtration does not reflect the true MW. FIGURE 8 illustrates the substrte specificity of PEG-Modified UK's.

Example 26 - EFFECT OF UK INHIBITORS ON THE ACTIVITY OF
~ATIVE UK AND L-MD.
It is known that in human plasma, protease inhibitors play an important role in retaining blood fluidity. Physiological activity of native UK when administered intraveneously is rapidly retarded not only by the interaction with these inhibitors but also by the fragmentation. The effect of UK inhibitors on the UK activity was examined using Fl, inhibitor fraction from human plasma, and placental UK
inhibitor. In the first experiment, U~s were incubated wieh the former at 37 then residual UK was assayed with S-2444. In the next experiment, placental UK

-:^

771~

inhibitor of different dilution was added to UKs then residual UK was assayed with a combination of plasminogen and S-2251.
In both cases, L-Md retained its activity better than native UK did. Neutral and chemically inert PEG
chains seem to protect UK from the action of UK
inhibitors. FIGURE 9 shows the stability of modified Urokinase to UK-Inhibitors in Human Plasma. ~IGURE 9b shows the effect of placental UK-Inhibitor on the Urokinase activity.

Example 27 - EXTENSION OF CIRCULATING LIVES OF UKs IN RABBITS
The short half-life (~/2) of native UK in the circulatory system often becomes a limitation of UK
therapy. As the ~20f native UK does not exceed several minutes, prolonged drop infusion is necessary to maintain the high plasmin level. Half lives of native and modified UKs were measured in rabbits by an injection of one of them followed by the determination of UK activity in the plasma drawn, periodically, from an ear vein.
As a re~ult, the half-life of native UK in the first phase was calculated as 4-5 minutes, L-Md as 30-40 minutes, M-Md as 80-100 minutes and H-Md as 110-150 minutes. ~20f modified UKs were extended about 10 to 30 times that of the native UK depending upon the 77~

respective degree of modification. In the case of M-Md or H-Md, a one compartment model is approximate enough to describe the pharmacokinetics, which suggests tha~
distribution to a second compartment is suppressed.
FIGURE 10 shows the UK activity of L-Md, M-Md and H-Md in Plasma after UK injection.

Example 28 - BEHAVIOR O~ 125I-NA'rIVE UK AND 125I-M-MD
IN RATS
Native UK and M-Md were labeled with 125I. Then, their ~2in the circulatory system and their distribution among organs were investigated in rats.
Extension of ~2f UK by PEG modification was also observed and was more prominent in these experiments.
Rapid decay of radioactivity in blood of rats injected with 125I-native UK was observed. On the contrary, radioactivity was retained well with 125I-M-Md. In the first phase, ~20f native UK was 1.1 min. while that of M-Md was 89.9 min.
Native UK administered intravenously accumulates in the liver and kidneys and other organs resulting in a short ~/2~ With M-Md, accumulation in these organs was suppressed probably due to less interaction of the M-Md molecule~ with organ cells because of the neutral and inert PEG chains on UK.
Tables 1 and 2 ~how the relative radioactivity in various tissues after 125I-native UK and 125I-M-Md ~L77~

injection, respectively. FIGURE 11 illustrates the relative radioactivity in Blood (~) for 125I-M-Md and 125I-native UK-RELATIVE RADIOACTIVITY AF~ER
125I-NATIVE UK INJ_CTIO~

Time Organ 5 min.10 min.20 min. 40 min.12 hr Brain 0.050.03 0.04 0.010.01 Lung 0.470.62 0.31 0.100.01 Thymus 0.030.02 0.03 0.030.01 Liver 27.8932.60 23.06 2.590.46 Heart 0.140.26 0.17 0.170.10 Kidney 3.686.82 2.40 0.400.25 Adrenal 0.05 -0.07 0.05 0.020.00 Spleen 0.39 1.32 0.67 0.050.02 RELATIVE RADIOACTIVITY AFTER
125I_M_MD INJECTION __ Time organ 5 min. 10 min.20 min~40 min.12 hr Brain 0.17 0.20 0.20 0.220.11 Lung 1.24 2.30 1.31 1.370.71 Thymus 0.15 0.21 0.15 0.240.11 Liver 6.84 8.7511.55 7.472.91 Heart 0.93 1.73 1.38 1.020.53 Kidney 2.21 3~10 3.83 3.270.68 Adrenal 0.06 0.07 0.08 0.060.04 Spleen 0.45 0.40 0.63 0.510.21 EXamP1e 29 - IMMUNOGENICITY AND TOXICITY OF M-MD
The changes in immunogenicity ~y chemical modification of proteins are of interest.

7~3 Immunogenicity of M-Md was compared with that of native UK by ASA reaction, Schultz-Dale test, PCA reaction and Ouchterlony method. Both native UK and M-Md were immunogenic for guinea pigs by ASA reaction after sensitization of them with the respective UK and FCA.
However, the immunological response to M-Md was weaker than that of native UK in the other three tests.
The results summarized in FIGURES 12a and 12b suggest that immunological determinants in M-Md are common with those in native UK. Furthermore, antigen production and/or reactivity with antigen are/is suppressed in M-Md compared with native UK. As native UK itself does not cause immunological response in man, M-Md is not expected to either.
The toxicity of M-Md was found to be extremely low. None of the mice died even after a shot of M-MD
at a dosage of 1,000,000 ~/kg nor afte~ repetitive injection of 500,000 ~/kg/day of M-Md.
FIGURE 12a shows a comparison of the immunogenicity of M-Md with that oE native UK by Schultz-Dale test~ FIGURE 12b shows a comparison of the immunogenicity of M-Md with that of native UK by Passive Cu~aneous Anaphylaxis (PCA) reaction.
In ~ummary, the instability of native UK has created a serious problem in formulating it for clinical purposes. The foregoing examples illustrate that covalent attachment of PEG chains to native UK
greatly increases resistance to inhibition of the UK by UK inhibitors. Moreover, covalent attachment of PEG
chains to native UK also prolonged ~2f the UK in the circulatory system. It seems likely that the PEG
chains may surround the UK molecule to form, in effect an inert capsule to protect the UK molecule from inhibitors and other proteases.
In the next set of examples attention was focused on _ vivo experiments using dogs, where the thrombolytic ability of PEG-modified UK (PEG-UK) was compared wlth that of native UK. In these examples, the PEG-UK used is the same as M-Md in Examples 25-29. The unit for UK activity was determined by the standard fibrin plate (F.P.) method. Plasmin was obtained from Sigma Chemical Co. FDP latex for dog FDP
was obtained from MBL Co., Ltd. Terufus~on~, a blood transfusion set type 1, obtained from Terumo Co., Ltd.
was connected with a three-way stopcock of type PX2-50 from Top Co., Ltd. using ATOM 6 Fr extension tubing.
Other materials used in the following examples are the same as used in Examples 25-29.
T~e haemostatic index for plasmin inhibitor, Fl, was determined by an addition of plasmin to the Fl fraction followed by the assay of residual pla~min with S-2251. F3, plasminogen, was determined with a ~Z~,t - 7 ~

combination of UK and S-2251. FDP was determined wi~h latex sensiti~ed with antigen to dog fibrinogen.
For the shun~ preparation, a dog was anaesthetized with ketamine hydrochloride then A. femoralis and V.
femoralis were exposed in the inguinal region. A
saline solution-filled shunt with the three-way stopcock was connected between them. Blood was allowed to run by opening the stopcock while monitoring the flow with a magnetic bloodflow meter. Either native UK
or PEG-UK dissolved in saline solution was administered from the stopcock. To maintain the anaesthesia, sodium barbiturate was injected.
For the preparation of artifical thrombus, A.
fermoralis of an anaesthetized dog was exposed by the same procedure. A transfusion catheter was fixed at a branch of A. femoralis, then the distal portion of the A. femoralis was clamped with two b~lldo~ clamps at a distance of 2 cm from each other and was emptied, then washed with saline solution. Dry oxygen was passed therethrough at a rate of 2 l/min for 10 minutes to injure intima. After injection of a fibrinogen solution followed by thrombin solution, the formed thrombus was allowed to age for 10 minutes. Then, the bulldog clamps were removed. Thrombus formation was confirmed angiographically by an injection of 10 ml of 60% Urografin~ from the catheter. UK dissolved in 16~
- ~o-saline was injected from the same catheter and angiographies were taken periodically thereafter.

Example 30 - CHANGES IN Fl, F3 AND FDP AFTER UK
INJECTION
Changes in the haemostatic indices are good indications of UK treatment. Often used are Fl (plasmin inhibi~or), F3 (plasminogen) and FDP (fibrin and/or fibrinogen degradation product). Native UK or PEG-UK was injected into a dog at the superficial vein of the fore leg at dosages shown in FIGURES 13a, b and c. Blood was sampled from the opposite side at certain time intervals and the Fl, F3 and FDP contents of the samples were determined.
A decrease in both Fl and F3 levels is a direct proof of plasminogen activation. The consumption of F
and F3 indicates that plasmin activated by UK from plasminogen (F3) was deactivated by its inhibitor (Fl) and was removed from the circulatory system. Another proof is a prominent increase in FDP. In a dog injected with PEG-UK, the FDP level was kept higher for several hours although its enæymatic activity to activate plasminogen was lower than that of native UK. These findings reveal clearly that chemical modification with PEG amplifies the physiological activity of UK.

7'~

FIGURE 13a illustrates a comparison of the Plasmin Inhibitor in Fl from Citrated Dog Plasma at certain time intervals after injection of native UK or PEG-UK
into a dog.
FIGURE 13b illustrates a comparison of the Plasminogen in F3 from Citrated Dog Plasma at certain time intervals after injection of native UK or PEG-UK
in~o a dog.
FIGURE 13c illustrates a comparison of the FDP in Dog Serum at certain time intervals after i.v.
injection of UK or PEG-UK into a dog.

Example 31 - THROMBOLYTIC EFFECT OF PEG-UK I~
EXPERIMENTAL THROMBOSIS
Thrombolytic activities of native UK and PEG-UK
were compared in dogs with experimental thrombosis. A
shunt having a fine filter was fixed between A.
femoralis and V. femoralis and then blood was run through it. After a complete stop of flow (several minutes) either native UK or PEG-UK was administered through the three-way stopcock at a dose of 10,000 ~/kg. As a result, the flow of blood in the dog with PEG-UK recovered while not in the dog having native UK administered even after repeated flushes of saline ~olution.

1' ;LI'i7.~3 -~2-Example 32 - INHIBITION OF THROMBUS FORMATION WITH
PEG-UK
The inhibitory effect of PEG-UK on the thrombus formation was investigated as compared with native UK
with a saline solution-filled shunt prepared by the same procedures de~cribed above. In this experiment either native UK or P~G-UK was administered prior to the blood circulation. The dose was 20,000 ~/kg in both cases. Results obtained are as follows:

1) In the dog without UK, the blood flow stopped within 5 minutes.
2) In the dog with native UK, the blood flow stopped within 5 minutes.
3) In the dog with PEG-UK, the blood flow continued during the experiment for >1 hour.
At this stage, the three filters were washed with saline. Complete inhibition of thrombus formation was observed in PEG-UK, which is probably due to the thrombin inhibition by FDP.

Example 33 - LYSIS OF ARTIFICIAL THROM~US WITH PEG-UK
USING ANGIOGRAM
The thombolytic effect of PEG-UK was evaluated in dogs bearing artifical thrombus in A. femoralis. After confirmation of thrombus formation, either native UK
(dog "A" ) or PEG-UK (dog "B") was injected from the transfusion catheter fixed on a proximal branch of the ~7~

thrombus at a dose of 10,00~ ~/kg. Angiographies were taken periodically to observe ~he recovery of blood flow. It is concluded from these angiographies that PEG-UK dissolved the artifical ~hrombus formed in A.
femoralis while native UK was unable to do so. FIGURE
14 illustrates a comparison of FDP levels in dogs bearing artifical thrombus in A. femoralis. Dog A had native UK injected into a proximal branch of the thrombus, while dog B had PEG-UK injected into the same.

Example 34 - INHIBITORY EFFECT ON THROMBUS FORMATION
Using a dog, a shunt was established under anesthesia by a blood transfu~ion filter ("Terufusion"
blood transfusion Set Type-l) between A. femoralis and V. femoralis and a blood flowmeter ("MF-26", rectangular wave electromagnetic bl~od f~-owmeter manufactured by Nippon Koden K.K.) was connected at the vein side. The filter had in advance been filled with physiological saline and, after administration of a drug, the blood was caused to flow through the shunt by opening a three-way stopcock. Without administration of the drug, thrombus was formed in the filter within 4-5 minutes after the blood had started flowing through the filter, whereby any further flow of the blood ~hrough the shunt was preven~ed. The time period until 1~ o 7 ~

the stoppage of the blood flow due to the formation of thrombus was not prolonged at all even when 200,000 units of unmodified urokinase were administered. On the contrary, thrombus was not formed nor was the blood flow rate reduced at all even after 40 minutes after the initiation of the blood flow through the shunt when PEG-DCT-UK, which pertains to the present invention, had been administered.
In summary, Examples 30-34 dealt with the thrombolysis by PEG-UK wherein the thrombolytic ability thereof was compared with that of native UK using two thrombosis models. The superiority of PEG-UK to native UK with respect to fibrinolytic activation is due to the PEG chains which appear to protect the UK molecule from deactivating interactions with inhibitors. This protection serves to extend the UK circulating life.
Having fully described the pre5ent-~nvention, it will be apparent to one of ordinary skill in the art that many changes and modifications can be made thereto without departing from the spirit or scope of the invention as set forth herein.

Claims (5)

THE EMBODIMENTS OF THE INVENTION IN WHICH AN EXCLUSIVE
PROPERTY OF PRIVILEGE IS CLAIMED ARE DEFINED AS FOLLOWS.
1. A method of extending the circulating life of a plasminogen activator in a mammalian bloodstream which comprises chemically modifying said plasminogen activator by bonding at least one polyalkylene glycol moiety there-to by means of a coupling agent to the amino acid side chains of said plasminogen activator.
2. The method of Claim 1, wherein said plasminogen activator is urokinase.
3. The method of Claim 1, wherein said mammalian bloodstream is a human bloodstream.
4. The method of Claim 1, wherein said polyalkylene glycol moiety is a methoxypolyethylene glycol moiety.
5. The method of Claim 4, wherein said methoxy-polyethylene glycol has a molecular weight of about 5,000.
CA000443230A 1981-10-30 1983-12-14 Plasminogen activator derivatives Expired CA1217718A (en)

Priority Applications (11)

Application Number Priority Date Filing Date Title
JP56172908A JPS5896026A (en) 1981-10-30 1981-10-30 Novel urokinase derivative, its preparation and thrombolytic agent containing the same
BR8206347A BR8206347A (en) 1981-10-30 1982-10-27 PROCESS TO PRODUCE PLASMINOGEN ACTIVATING DERIVATIVES
US06/437,009 US4495285A (en) 1981-10-30 1982-10-27 Plasminogen activator derivatives
CH6304/82A CH658669A5 (en) 1981-10-30 1982-10-28 DERIVATIVES OF PLASMINOGEN ACTIVATORS.
DE19823240174 DE3240174A1 (en) 1981-10-30 1982-10-29 Stable plasminogen activator derivs. - contg. poly:alkylene glycol linked to side chains
FR8218223A FR2515684B1 (en) 1981-10-30 1982-10-29 NEW DERIVATIVE OF A NON-IMMUNOGENIC PLASMINOGEN ACTIVATOR OF HUMAN ORIGIN
CA000414556A CA1203764A (en) 1981-10-30 1982-10-29 Plasminogen activator derivatives
GB08230987A GB2110219B (en) 1981-10-30 1982-10-29 Plasminogen activator derivatives
SE8206173A SE457800B (en) 1981-10-30 1982-10-29 PLASMINOGENAKTIVATORDERIVAT
US06/546,590 US4640835A (en) 1981-10-30 1983-10-28 Plasminogen activator derivatives
CA000443230A CA1217718A (en) 1981-10-30 1983-12-14 Plasminogen activator derivatives

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
JP56172908A JPS5896026A (en) 1981-10-30 1981-10-30 Novel urokinase derivative, its preparation and thrombolytic agent containing the same
US06/546,590 US4640835A (en) 1981-10-30 1983-10-28 Plasminogen activator derivatives
CA000443230A CA1217718A (en) 1981-10-30 1983-12-14 Plasminogen activator derivatives

Publications (1)

Publication Number Publication Date
CA1217718A true CA1217718A (en) 1987-02-10

Family

ID=27167396

Family Applications (1)

Application Number Title Priority Date Filing Date
CA000443230A Expired CA1217718A (en) 1981-10-30 1983-12-14 Plasminogen activator derivatives

Country Status (2)

Country Link
US (1) US4640835A (en)
CA (1) CA1217718A (en)

Families Citing this family (781)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB8430252D0 (en) * 1984-11-30 1985-01-09 Beecham Group Plc Compounds
US4791192A (en) * 1986-06-26 1988-12-13 Takeda Chemical Industries, Ltd. Chemically modified protein with polyethyleneglycol
US5045190A (en) * 1988-11-08 1991-09-03 Carbonell Ruben G Chromatography apparatus
US5166322A (en) * 1989-04-21 1992-11-24 Genetics Institute Cysteine added variants of interleukin-3 and chemical modifications thereof
JPH05501855A (en) * 1989-05-17 1993-04-08 リサーチ、コーポレーション、テクノロジーズ、インコーポレーテッド Methods and compositions for treating thrombosis in mammals
FR2661920B1 (en) * 1990-05-10 1995-10-06 Serbio METHOD FOR DETERMINING A PLASMINOGEN ACTIVATOR AND ITS INHIBITOR.
US5595732A (en) * 1991-03-25 1997-01-21 Hoffmann-La Roche Inc. Polyethylene-protein conjugates
US5272076A (en) * 1991-06-13 1993-12-21 Eli Lilly And Company Compounds and methods for treatment of thromboembolic disorders using an adduct of t-PA
US5382657A (en) * 1992-08-26 1995-01-17 Hoffmann-La Roche Inc. Peg-interferon conjugates
US5877016A (en) 1994-03-18 1999-03-02 Genentech, Inc. Human trk receptors and neurotrophic factor inhibitors
US6458762B1 (en) 1994-03-28 2002-10-01 Baxter International, Inc. Therapeutic use of hemoglobin for preserving tissue viability and reducing restenosis
US5795569A (en) * 1994-03-31 1998-08-18 Amgen Inc. Mono-pegylated proteins that stimulate megakaryocyte growth and differentiation
HU218893B (en) * 1994-03-31 2000-12-28 Amgen Inc. Water soluble compositions and methods for stimulating megakaryocyte growth and differentiation
US5708142A (en) 1994-05-27 1998-01-13 Genentech, Inc. Tumor necrosis factor receptor-associated factors
DE4423131A1 (en) * 1994-07-01 1996-01-04 Bayer Ag New hIL-4 mutant proteins as antagonists or partial agonists of human interleukin 4
MX9704137A (en) * 1994-12-07 1997-09-30 Novo Nordisk As Polypeptide with reduced allergenicity.
EA001220B1 (en) 1995-06-07 2000-12-25 Глаксо Груп Лимитед Peptides or peptidemimetics bound to thrombopoietin receptor, pharmaceutical composition and method of treatment
US5869451A (en) 1995-06-07 1999-02-09 Glaxo Group Limited Peptides and compounds that bind to a receptor
BR9510676A (en) * 1995-12-29 1999-11-23 Procter & Gamble Detergent compositions comprising immobilized enzymes
US6998116B1 (en) * 1996-01-09 2006-02-14 Genentech, Inc. Apo-2 ligand
US6046048A (en) * 1996-01-09 2000-04-04 Genetech, Inc. Apo-2 ligand
US6030945A (en) * 1996-01-09 2000-02-29 Genentech, Inc. Apo-2 ligand
US20050089958A1 (en) * 1996-01-09 2005-04-28 Genentech, Inc. Apo-2 ligand
US20020165157A1 (en) * 1996-04-01 2002-11-07 Genentech, Inc. Apo-2LI and Apo-3 polypeptides
US6469144B1 (en) 1996-04-01 2002-10-22 Genentech, Inc. Apo-2LI and Apo-3 polypeptides
US7091311B2 (en) * 1996-06-07 2006-08-15 Smithkline Beecham Corporation Peptides and compounds that bind to a receptor
US6159462A (en) * 1996-08-16 2000-12-12 Genentech, Inc. Uses of Wnt polypeptides
US5851984A (en) * 1996-08-16 1998-12-22 Genentech, Inc. Method of enhancing proliferation or differentiation of hematopoietic stem cells using Wnt polypeptides
US6462176B1 (en) * 1996-09-23 2002-10-08 Genentech, Inc. Apo-3 polypeptide
AU725133B2 (en) * 1997-01-15 2000-10-05 Polaris Group Modified tumor necrosis factor
ATE356212T1 (en) 1997-01-31 2007-03-15 Genentech Inc O-FUKOSYL TRANSFERASE
US20020102706A1 (en) * 1997-06-18 2002-08-01 Genentech, Inc. Apo-2DcR
ES2236634T3 (en) 1997-04-07 2005-07-16 Genentech, Inc. ANTI-VEGF ANTIBODIES.
KR100794454B1 (en) 1997-04-07 2008-01-16 제넨테크, 인크. Anti-VEGF Antibodies
US20100152426A1 (en) * 1997-05-15 2010-06-17 Ashkenazi Avi J Apo-2 receptor fusion proteins
US6342369B1 (en) * 1997-05-15 2002-01-29 Genentech, Inc. Apo-2-receptor
JP2001511653A (en) 1997-05-15 2001-08-14 ジェネンテク,インコーポレイテッド Apo-2 receptor
CA2293724C (en) 1997-06-05 2010-02-02 Xiaodong Wang Apaf-1, the ced-4 human homolog, an activator of caspase-3
EP2083079A1 (en) * 1997-06-18 2009-07-29 Genentech, Inc. Apo-2DcR
AU751880B2 (en) * 1997-06-25 2002-08-29 Novozymes A/S A modified polypeptide
US6342220B1 (en) 1997-08-25 2002-01-29 Genentech, Inc. Agonist antibodies
US20030175856A1 (en) * 1997-08-26 2003-09-18 Genetech, Inc. Rtd receptor
DK1009817T3 (en) * 1997-08-26 2010-01-18 Genentech Inc RTD receptor
CA2382506A1 (en) 1997-09-17 1999-03-25 Genentech, Inc. Novel polypeptides and nucleic acids encoding pro293 which are useful for treating disorders of the pancreas
US20040231011A1 (en) * 2001-06-28 2004-11-18 Genentech, Inc. DcR3 polypeptide, a TNFR homolog
WO1999014330A1 (en) 1997-09-18 1999-03-25 Genentech, Inc. DcR3 POLYPEPTIDE, A TNFR HOMOLOG
IL135051A0 (en) 1997-10-10 2001-05-20 Genentech Inc Apo-3 ligand polypeptide
EP2033970A3 (en) 1997-10-29 2009-06-17 Genentech, Inc. Polypeptides and nucleic acids encoding the same
US6387657B1 (en) 1997-10-29 2002-05-14 Genentech, Inc. WISP polypeptides and nucleic acids encoding same
US7192589B2 (en) 1998-09-16 2007-03-20 Genentech, Inc. Treatment of inflammatory disorders with STIgMA immunoadhesins
EP2014677A1 (en) 1997-11-21 2009-01-14 Genentech, Inc. A-33 related antigens and their pharmacological uses
DK1045906T3 (en) 1998-01-15 2009-02-16 Genentech Inc APO-2 ligand
NZ525914A (en) 1998-03-10 2004-03-26 Genentech Inc Novel polypeptides and nucleic acids encoding the same
ES2389387T3 (en) 1998-03-17 2012-10-25 Genentech, Inc. Homologous VEGF and BMP1 polypeptides
EP1075511A1 (en) * 1998-05-08 2001-02-14 University Of Southern California Size enhanced fibrinolytic enzymes
EP3112468A1 (en) 1998-05-15 2017-01-04 Genentech, Inc. Il-17 homologous polypeptides and therapeutic uses thereof
EP2333069A3 (en) 1998-05-15 2011-09-14 Genentech, Inc. Therapeutic uses of IL-17 homologous polypeptides
EP1865061A3 (en) 1998-05-15 2007-12-19 Genentech, Inc. IL-17 homologous polypeptides and therapeutic uses thereof
ES2395693T3 (en) * 1998-06-12 2013-02-14 Genentech, Inc. Monoclonal antibodies, cross-reactive antibodies and process for manufacturing them
US20020172678A1 (en) 2000-06-23 2002-11-21 Napoleone Ferrara EG-VEGF nucleic acids and polypeptides and methods of use
DE69934425T2 (en) 1998-10-23 2007-09-27 Amgen Inc., Thousand Oaks THROMBOPOIETIN SUBSTITUTE
SI1135498T1 (en) 1998-11-18 2008-06-30 Genentech Inc Antibody variants with higher binding affinity compared to parent antibodies
AUPP785098A0 (en) * 1998-12-21 1999-01-21 Victor Chang Cardiac Research Institute, The Treatment of heart disease
EP2075335A3 (en) 1998-12-22 2009-09-30 Genentech, Inc. Methods and compositions for inhibiting neoplastic cell growth
JP5456222B2 (en) 1998-12-23 2014-03-26 ジェネンテック, インコーポレイテッド IL-1 related polypeptides
EP1792989A1 (en) 1999-04-12 2007-06-06 Agensys, Inc. 13 Transmembrane protein expressed in prostate cancer
CA2369413C (en) 1999-04-12 2013-07-09 Agensys, Inc. Transmembrane protein expressed in prostate and other cancers
US6635249B1 (en) 1999-04-23 2003-10-21 Cenes Pharmaceuticals, Inc. Methods for treating congestive heart failure
AU5152700A (en) 1999-06-15 2001-01-02 Genentech Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same
EP1200590B1 (en) 1999-08-12 2009-01-07 Agensys, Inc. C-type lectin transmembrane antigen expressed in human prostate cancer and uses thereof
US7459540B1 (en) * 1999-09-07 2008-12-02 Amgen Inc. Fibroblast growth factor-like polypeptides
WO2001025434A1 (en) 1999-10-05 2001-04-12 Agensys, Inc. G protein-coupled receptor up-regulated in prostate cancer and uses thereof
US7332275B2 (en) 1999-10-13 2008-02-19 Sequenom, Inc. Methods for detecting methylated nucleotides
US6893818B1 (en) * 1999-10-28 2005-05-17 Agensys, Inc. Gene upregulated in cancers of the prostate
KR20020056923A (en) 1999-11-18 2002-07-10 코르바스 인터내셔날, 인코포레이티드 Nucleic acids encoding endotheliases, endotheliases and uses thereof
US6703480B1 (en) 1999-11-24 2004-03-09 Palani Balu Peptide dimers as agonists of the erythropoientin (EPO) receptor, and associated methods of synthesis and use
CA2490853A1 (en) 1999-12-01 2001-06-07 Genentech, Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same
US7109299B1 (en) 1999-12-16 2006-09-19 Affymax, Inc. Peptides and compounds that bind to the IL-5 receptor
DK1897945T3 (en) 1999-12-23 2012-05-07 Genentech Inc IL-17 homologous polypeptides and therapeutic uses thereof.
ATE424457T1 (en) * 2000-01-13 2009-03-15 Genentech Inc HUMAN STRA6 POLYPEPTIDES
US7700341B2 (en) * 2000-02-03 2010-04-20 Dendreon Corporation Nucleic acid molecules encoding transmembrane serine proteases, the encoded proteins and methods based thereon
DK1255558T3 (en) 2000-02-16 2006-10-23 Genentech Inc Anti-April antibodies and hybridoma cells
US7101974B2 (en) 2000-03-02 2006-09-05 Xencor TNF-αvariants
US6740520B2 (en) 2000-03-21 2004-05-25 Genentech, Inc. Cytokine receptor and nucleic acids encoding the same
US20040086970A1 (en) * 2000-03-22 2004-05-06 Genentech, Inc. Novel cytokine receptors and nucleic acids encoding the same
US6667300B2 (en) 2000-04-25 2003-12-23 Icos Corporation Inhibitors of human phosphatidylinositol 3-kinase delta
DK2042597T3 (en) 2000-06-23 2014-08-11 Genentech Inc COMPOSITIONS AND PROCEDURES FOR DIAGNOSIS AND TREATMENT OF DISEASES INVOLVING ANGIOGENESIS
CA2709771A1 (en) 2000-06-23 2002-01-03 Genentech, Inc. Compositions and methods for the diagnosis and treatment of disorders involving angiogenesis
FR2811323B1 (en) * 2000-07-07 2006-10-06 Fuma Tech Gmbh HYBRID MATERIAL, USE OF SAID HYBRID MATERIAL, AND METHOD OF MANUFACTURING THE SAME
DE60136281D1 (en) 2000-08-24 2008-12-04 Genentech Inc METHOD FOR INHIBITING IL-22-INDUCED PAP1
PT1313850E (en) 2000-08-28 2008-11-18 Agensys Inc Nucleic acid and corresponding protein entitled 85p1b3 useful in treatment and detection of cancer
EP1944317A3 (en) 2000-09-01 2008-09-17 Genentech, Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same
US20030044803A1 (en) * 2000-09-22 2003-03-06 Pedersen Finn Skou Methods for diagnosis and treatment of diseases associated with altered expression of JAK1
US20020164576A1 (en) * 2000-09-22 2002-11-07 Pedersen Finn Skou Methods for diagnosis and treatment of diseases associated with altered expression of Nrf2
US20020115058A1 (en) * 2000-09-22 2002-08-22 Pedersen Finn Skou Methods for diagnosis and treatment of diseases associated with altered expression of Pik3r1
US6576452B1 (en) * 2000-10-04 2003-06-10 Genencor International, Inc. 2,5-diketo-L-gluconic acid reductases and methods of use
US6673580B2 (en) * 2000-10-27 2004-01-06 Genentech, Inc. Identification and modification of immunodominant epitopes in polypeptides
EP1355671A2 (en) * 2000-11-30 2003-10-29 Nektar Therapeutics Al, Corporation Water-soluble polymer conjugates of triazine derivatives
US20030165878A1 (en) * 2000-12-22 2003-09-04 Morris David W. Novel compositions and methods in cancer associated with altered expression of MCM3AP
US7700274B2 (en) * 2000-12-22 2010-04-20 Sagres Discovery, Inc. Compositions and methods in cancer associated with altered expression of KCNJ9
US20030087252A1 (en) * 2000-12-22 2003-05-08 Morris David W. Novel compositions and methods in cancer associated with altered expression of PRDM11
US20030099963A1 (en) * 2000-12-22 2003-05-29 Morris David W. Novel compositions and methods in cancer associated with altered expression of TBX21
US7820447B2 (en) 2000-12-22 2010-10-26 Sagres Discovery Inc. Compositions and methods for cancer
US7892730B2 (en) * 2000-12-22 2011-02-22 Sagres Discovery, Inc. Compositions and methods for cancer
US7645441B2 (en) 2000-12-22 2010-01-12 Sagres Discovery Inc. Compositions and methods in cancer associated with altered expression of PRLR
US20030232334A1 (en) 2000-12-22 2003-12-18 Morris David W. Novel compositions and methods for cancer
US7754208B2 (en) 2001-01-17 2010-07-13 Trubion Pharmaceuticals, Inc. Binding domain-immunoglobulin fusion proteins
US7829084B2 (en) * 2001-01-17 2010-11-09 Trubion Pharmaceuticals, Inc. Binding constructs and methods for use thereof
US20020123068A1 (en) * 2001-01-31 2002-09-05 Dwyer Brian P. Water-soluble, fluorescent, & electrophoretically mobile peptidic substrates for enzymatic reactions and methods for their use in high-throughput screening assays
US7087726B2 (en) 2001-02-22 2006-08-08 Genentech, Inc. Anti-interferon-α antibodies
US6924358B2 (en) 2001-03-05 2005-08-02 Agensys, Inc. 121P1F1: a tissue specific protein highly expressed in various cancers
US7125703B2 (en) 2001-03-13 2006-10-24 Dendreon Corporation Nucleic acid molecules encoding a transmembrane serine protease 7, the encoded polypeptides and methods based thereon
US7271240B2 (en) 2001-03-14 2007-09-18 Agensys, Inc. 125P5C8: a tissue specific protein highly expressed in various cancers
KR20040011480A (en) 2001-03-22 2004-02-05 덴드레온 샌 디에고 엘엘씨 Nucleic acid molecules encoding serine protease CVSP14, the encoded polypeptides and methods based thereon
NZ527971A (en) 2001-03-27 2006-03-31 Dendreon Corp Nucleic acid molecules encoding a transmembrane serine protease 9, the encoded polypeptides and methods based thereon
WO2002083921A2 (en) 2001-04-10 2002-10-24 Agensys, Inc. Nuleic acids and corresponding proteins useful in the detection and treatment of various cancers
US20030191073A1 (en) 2001-11-07 2003-10-09 Challita-Eid Pia M. Nucleic acid and corresponding protein entitled 161P2F10B useful in treatment and detection of cancer
JP2005506832A (en) 2001-05-14 2005-03-10 デンドレオン・サンディエゴ・リミテッド・ライアビリティ・カンパニー Nucleic acid molecule encoding transmembrane serine protease 10, encoded polypeptide and method based thereon
US20070160576A1 (en) 2001-06-05 2007-07-12 Genentech, Inc. IL-17A/F heterologous polypeptides and therapeutic uses thereof
US20060270003A1 (en) 2003-07-08 2006-11-30 Genentech, Inc. IL-17A/F heterologous polypeptides and therapeutic uses thereof
CA2633171C (en) 2001-06-20 2012-11-20 Genentech, Inc. Antibodies against tumor-associated antigenic target (tat) polypeptides
JP2005510208A (en) * 2001-08-03 2005-04-21 ジェネンテック・インコーポレーテッド TACIs and BR3 polypeptides and uses thereof
HU230373B1 (en) 2001-08-29 2016-03-29 Genentech Inc Bv8 nucleic acids and polypeptides with mitogenic activity
US20040235068A1 (en) * 2001-09-05 2004-11-25 Levinson Arthur D. Methods for the identification of polypeptide antigens associated with disorders involving aberrant cell proliferation and compositions useful for the treatment of such disorders
ES2537074T3 (en) 2001-09-06 2015-06-02 Agensys, Inc. Nucleic acid and corresponding protein called STEAP-1 useful in the treatment and detection of cancer
NZ573831A (en) 2001-09-18 2010-07-30 Genentech Inc Compositions and methods for the diagnosis and treatment of tumor, particularly breast tumor - TAT193
US20070098728A1 (en) * 2001-09-24 2007-05-03 Pedersen Finn S Novel compositions and methods in cancer
US7320789B2 (en) 2001-09-26 2008-01-22 Wyeth Antibody inhibitors of GDF-8 and uses thereof
US7138370B2 (en) 2001-10-11 2006-11-21 Amgen Inc. Specific binding agents of human angiopoietin-2
US7521053B2 (en) 2001-10-11 2009-04-21 Amgen Inc. Angiopoietin-2 specific binding agents
US20040126762A1 (en) * 2002-12-17 2004-07-01 Morris David W. Novel compositions and methods in cancer
US20040166490A1 (en) * 2002-12-17 2004-08-26 Morris David W. Novel therapeutic targets in cancer
WO2003044179A2 (en) * 2001-11-20 2003-05-30 Dendreon San Diego Llc Nucleic acid molecules encoding serine protease 17, the encoded polypeptides and methods based thereon
US20040180344A1 (en) * 2003-03-14 2004-09-16 Morris David W. Novel therapeutic targets in cancer
US20060040262A1 (en) * 2002-12-27 2006-02-23 Morris David W Novel compositions and methods in cancer
US20040197778A1 (en) * 2002-12-26 2004-10-07 Sagres Discovery, Inc. Novel compositions and methods in cancer
WO2003050276A1 (en) * 2001-12-05 2003-06-19 Dow Global Technologies Inc. Method for immobilizing a biologic in a polyurethane-hydrogel composition, a composition prepared from the method, and biomedical applications
NZ533933A (en) 2002-01-02 2008-06-30 Genentech Inc Compositions and methods for the diagnosis and treatment of glioma tumor
KR20040096592A (en) 2002-02-21 2004-11-16 와이어쓰 Follistatin domain containing proteins
EP1575480A4 (en) 2002-02-22 2008-08-06 Genentech Inc Compositions and methods for the treatment of immune related diseases
US7303896B2 (en) 2002-02-25 2007-12-04 Genentech, Inc. Nucleic acid encoding novel type-1 cytokine receptor GLM-R
US20100311954A1 (en) * 2002-03-01 2010-12-09 Xencor, Inc. Optimized Proteins that Target Ep-CAM
US20070122406A1 (en) 2005-07-08 2007-05-31 Xencor, Inc. Optimized proteins that target Ep-CAM
US20040023267A1 (en) * 2002-03-21 2004-02-05 Morris David W. Novel compositions and methods in cancer
CA2481507A1 (en) 2002-04-16 2003-10-30 Genentech, Inc. Compositions and methods for the diagnosis and treatment of tumor
CA2484676A1 (en) * 2002-05-03 2003-11-13 Sequenom, Inc. Kinase anchor protein muteins, peptides thereof, and related methods
US7351542B2 (en) 2002-05-20 2008-04-01 The Regents Of The University Of California Methods of modulating tubulin deacetylase activity
US20040001801A1 (en) * 2002-05-23 2004-01-01 Corvas International, Inc. Conjugates activated by cell surface proteases and therapeutic uses thereof
WO2003099320A1 (en) 2002-05-24 2003-12-04 Zensun (Shanghai) Sci-Tech.Ltd Neuregulin based methods and compositions for treating viral myocarditis and dilated cardiomyopathy
EP2305710A3 (en) 2002-06-03 2013-05-29 Genentech, Inc. Synthetic antibody phage libraries
AU2003243400B2 (en) * 2002-06-07 2009-10-29 Genentech, Inc. Compositions and methods for the diagnosis and treatment of tumor
CA2489588A1 (en) 2002-07-08 2004-01-15 Genentech, Inc. Compositions and methods for the treatment of immune related diseases
WO2004011611A2 (en) * 2002-07-25 2004-02-05 Genentech, Inc. Taci antibodies and uses thereof
MXPA05001364A (en) * 2002-08-02 2005-08-03 Wyeth Corp Mk2 interacting proteins.
EP1546316B1 (en) * 2002-08-12 2010-12-01 Danisco US Inc. Mutant e. coli appa phytase enzymes and natural variants thereof, nucleic acids encoding such phytase enzymes, vectors and host cells incorporating same and methods of making and using same
WO2004016733A2 (en) 2002-08-16 2004-02-26 Agensys, Inc. Nucleic acid and corresponding protein entitled 251p5g2 useful in treatment and detection of cancer
AU2003265361A1 (en) * 2002-08-28 2004-03-19 Pharmacia Corporation Stable ph optimized formulation of a modified antibody
WO2004019860A2 (en) * 2002-08-28 2004-03-11 Pharmacia Corporation Formulations of modified antibodies and methods of making the same
AU2003276874B2 (en) 2002-09-11 2009-09-03 Genentech, Inc. Novel compositions and methods for the treatment of immune related diseases
EP2277532A1 (en) 2002-09-11 2011-01-26 Genentech, Inc. Novel composition and methods for the treatment of immune related diseases
CA2498274A1 (en) 2002-09-16 2004-03-25 Genentech, Inc. Compositions and methods for the diagnosis of immune related diseases using pro7
BR0314591A (en) 2002-09-18 2005-08-09 Ortho Mcneil Pharm Inc Platelet augmentation methods and hematopoietic stem cell production
EP1585482A4 (en) 2002-09-25 2009-09-09 Genentech Inc Nouvelles compositions et methodes de traitement du psoriasis
US20040149235A1 (en) * 2002-10-04 2004-08-05 Pogue Albert S. Apparatus and method for removal of waste from animal production facilities
WO2004039956A2 (en) 2002-10-29 2004-05-13 Genentech, Inc. Compositions and methods for the treatment of immune related diseases
WO2004110345A2 (en) * 2002-10-29 2004-12-23 Pharmacia Corporation Differentially expressed genes involved in cancer, the polypeptides encoded thereby, and methods of using the same
JP2006516094A (en) 2002-11-08 2006-06-22 ジェネンテック・インコーポレーテッド Compositions and methods for treatment of natural killer cell related diseases
EP2308968A1 (en) 2002-11-26 2011-04-13 Genentech, Inc. Compositions and methods for the treatment of immune related diseases
AU2002352976B2 (en) 2002-11-27 2007-11-08 Agensys, Inc. Nucleic acid corresponding protein entitled 24P4C12 useful in treatment and detection of cancer
US7553930B2 (en) * 2003-01-06 2009-06-30 Xencor, Inc. BAFF variants and methods thereof
US20050130892A1 (en) * 2003-03-07 2005-06-16 Xencor, Inc. BAFF variants and methods thereof
US20060014248A1 (en) * 2003-01-06 2006-01-19 Xencor, Inc. TNF super family members with altered immunogenicity
US20050221443A1 (en) * 2003-01-06 2005-10-06 Xencor, Inc. Tumor necrosis factor super family agonists
EP1581904A2 (en) * 2003-01-08 2005-10-05 Xencor, Inc. Novel proteins with altered immunogenicity
CA2860151A1 (en) 2003-02-10 2004-08-26 Agensys, Inc. Nucleic acid and corresponding protein named 158p1d7 useful in the treatment and detection of bladder and other cancers
EP1592708A2 (en) 2003-02-14 2005-11-09 Sagres Discovery, Inc. Therapeutic gpcr targets in cancer
US20070218071A1 (en) * 2003-09-15 2007-09-20 Morris David W Novel therapeutic targets in cancer
US20040170982A1 (en) 2003-02-14 2004-09-02 Morris David W. Novel therapeutic targets in cancer
US7767387B2 (en) * 2003-06-13 2010-08-03 Sagres Discovery, Inc. Therapeutic targets in cancer
KR101118340B1 (en) 2003-03-12 2012-04-12 제넨테크, 인크. Use of bv8 and/or eg-vegf to promote hematopoiesis
JP4914209B2 (en) * 2003-03-14 2012-04-11 ワイス Antibody against human IL-21 receptor and use of the antibody
MXPA05010411A (en) * 2003-03-28 2006-05-31 Biopolymed Inc Biologically active material conjugated with biocompatible polymer with 1:1 complex, preparation method thereof and pharmaceutical composition comprising the same.
US20050025763A1 (en) 2003-05-08 2005-02-03 Protein Design Laboratories, Inc. Therapeutic use of anti-CS1 antibodies
US7709610B2 (en) 2003-05-08 2010-05-04 Facet Biotech Corporation Therapeutic use of anti-CS1 antibodies
PT1629088E (en) 2003-05-30 2012-04-10 Agensys Inc Prostate stem cell antigen (psca) variants and subsequences thereof
UA101945C2 (en) 2003-05-30 2013-05-27 Дженентек, Инк. Treatment of cancer using bevacizumab
PT1631313E (en) * 2003-06-05 2015-07-02 Genentech Inc Combination therapy for b cell disorders
US20050163775A1 (en) * 2003-06-05 2005-07-28 Genentech, Inc. Combination therapy for B cell disorders
US7939058B2 (en) 2003-07-03 2011-05-10 University Of Southern California Uses of IL-12 in hematopoiesis
AR046071A1 (en) 2003-07-10 2005-11-23 Hoffmann La Roche ANTIBODIES AGAINST RECEIVER I OF THE INSULINAL TYPE GROWTH FACTOR AND THE USES OF THE SAME
KR20110129988A (en) 2003-07-18 2011-12-02 암젠 인코포레이티드 Specific binding agents to hepatocyte growth factor
WO2005019258A2 (en) 2003-08-11 2005-03-03 Genentech, Inc. Compositions and methods for the treatment of immune related diseases
WO2005016349A1 (en) * 2003-08-14 2005-02-24 Icos Corporation Methods of inhibiting leukocyte accumulation
US20050043239A1 (en) * 2003-08-14 2005-02-24 Jason Douangpanya Methods of inhibiting immune responses stimulated by an endogenous factor
AR045614A1 (en) * 2003-09-10 2005-11-02 Hoffmann La Roche ANTIBODIES AGAINST THE RECEIVER OF INTERLEUQUINA- 1 AND USES OF THE SAME
US8883147B2 (en) 2004-10-21 2014-11-11 Xencor, Inc. Immunoglobulins insertions, deletions, and substitutions
US8399618B2 (en) 2004-10-21 2013-03-19 Xencor, Inc. Immunoglobulin insertions, deletions, and substitutions
US20060134105A1 (en) * 2004-10-21 2006-06-22 Xencor, Inc. IgG immunoglobulin variants with optimized effector function
US20070281896A1 (en) * 2003-09-30 2007-12-06 Morris David W Novel compositions and methods in cancer
WO2005035569A2 (en) * 2003-10-10 2005-04-21 Five Prime Therapeutics, Inc. Kiaa0779, splice variants thereof, and methods of their use
WO2005035564A2 (en) 2003-10-10 2005-04-21 Xencor, Inc. Protein based tnf-alpha variants for the treatment of tnf-alpha related disorders
PT2161283E (en) 2003-11-17 2014-08-29 Genentech Inc Compositions comprising antibodies against cd79b conjugated to a growth inhibitory agent or cytotoxic agent and methods for the treatment of tumor of hematopoietic origin
HUE026132T2 (en) 2004-01-07 2016-05-30 Novartis Vaccines & Diagnostics Inc M-csf-specific monoclonal antibody and uses thereof
US20050169970A1 (en) * 2004-02-02 2005-08-04 Unilever Bestfoods, North America Food composition with fibers
AU2005230848B9 (en) * 2004-03-31 2011-06-02 Genentech, Inc. Humanized anti-TGF-beta antibodies
US7794713B2 (en) * 2004-04-07 2010-09-14 Lpath, Inc. Compositions and methods for the treatment and prevention of hyperproliferative diseases
EP2067789A1 (en) 2004-04-13 2009-06-10 F. Hoffmann-La Roche Ag Anti-P selectin antibodies
EP3943494A1 (en) * 2004-05-13 2022-01-26 Icos Corporation Quinazolinones as inhibitors of human phosphatidylinositol 3-kinase delta
JP2008500338A (en) * 2004-05-25 2008-01-10 イコス・コーポレイション Method for treating and / or preventing abnormal proliferation of hematopoietic cells
US7361493B1 (en) 2004-05-26 2008-04-22 The United States Of America As Represented By The Secretary Of The Department Of Veterans Affairs Production of urokinase in a three-dimensional cell culture
WO2005118864A2 (en) 2004-05-28 2005-12-15 Agensys, Inc. Antibodies and related molecules that bind to psca proteins
US20060014680A1 (en) * 2004-07-13 2006-01-19 Caiding Xu Peptides and compounds that bind to the IL-5 receptor
US20060024677A1 (en) 2004-07-20 2006-02-02 Morris David W Novel therapeutic targets in cancer
ES2339789T3 (en) 2004-07-20 2010-05-25 Genentech, Inc. PROTEIN 4 INHIBITORS OF ANGIOPOYETINE TYPE, COMBINATIONS AND ITS USE.
TWI309240B (en) 2004-09-17 2009-05-01 Hoffmann La Roche Anti-ox40l antibodies
AU2005286662B2 (en) 2004-09-23 2011-10-06 Vasgene Therapeutics, Inc. Polypeptide compounds for inhibiting angiogenesis and tumor growth
SI1797127T1 (en) * 2004-09-24 2017-09-29 Amgen Inc. Modified fc molecules
DE602005021534D1 (en) 2004-10-15 2010-07-08 Us Gov Health & Human Serv AMPHIPATHIC HELIXFUL MULTIDOMADE PEPTIDES AND METHOD FOR THE APPLICATION
US8802820B2 (en) 2004-11-12 2014-08-12 Xencor, Inc. Fc variants with altered binding to FcRn
WO2006053301A2 (en) 2004-11-12 2006-05-18 Xencor, Inc. Fc variants with altered binding to fcrn
JP2008521411A (en) 2004-11-30 2008-06-26 キュラジェン コーポレイション Antibodies against GPNMB and uses thereof
US20060134698A1 (en) * 2004-12-20 2006-06-22 Evanston Northwestern Healthcare Research Institute Methods for treating cardiac disease by modifying an N-terminal domain of troponin I
AU2006208226A1 (en) * 2005-01-24 2006-08-03 Amgen Inc. Humanized anti-amyloid antibody
CA2598409A1 (en) * 2005-02-17 2006-08-24 Icos Corporation Phosphoinositide 3-kinase inhibitors for inhibiting leukocyte accumulation
AU2006230563B8 (en) 2005-03-31 2010-06-17 Agensys, Inc. Antibodies and related molecules that bind to 161P2F10B proteins
TW200720289A (en) * 2005-04-01 2007-06-01 Hoffmann La Roche Antibodies against CCR5 and uses thereof
EP2062591A1 (en) 2005-04-07 2009-05-27 Novartis Vaccines and Diagnostics, Inc. CACNA1E in cancer diagnosis detection and treatment
EP2083088A3 (en) 2005-04-07 2009-10-14 Novartis Vaccines and Diagnostics, Inc. Cancer-related genes
US7592429B2 (en) 2005-05-03 2009-09-22 Ucb Sa Sclerostin-binding antibody
US8003108B2 (en) 2005-05-03 2011-08-23 Amgen Inc. Sclerostin epitopes
US20060271262A1 (en) * 2005-05-24 2006-11-30 Mclain Harry P Iii Wireless agricultural network
NZ563341A (en) 2005-06-06 2009-10-30 Genentech Inc Methods for identifying agents that modulate a gene that encodes for a PRO1568 polypeptide
US8389469B2 (en) * 2005-06-06 2013-03-05 The Rockefeller University Bacteriophage lysins for Bacillus anthracis
US8252756B2 (en) 2005-06-14 2012-08-28 Northwestern University Nucleic acid functionalized nanoparticles for therapeutic applications
US7582291B2 (en) * 2005-06-30 2009-09-01 The Rockefeller University Bacteriophage lysins for Enterococcus faecalis, Enterococcus faecium and other bacteria
CN105012953B (en) 2005-07-25 2018-06-22 阿普泰沃研发有限责任公司 B- cells are reduced with CD37- specificity and CD20- specific binding molecules
WO2007016240A2 (en) * 2005-07-28 2007-02-08 Novartis Ag Use of antibody to m-csf
EP1913027B1 (en) * 2005-07-28 2015-03-04 Novartis AG M-csf specific monoclonal antibody and uses thereof
US8008453B2 (en) 2005-08-12 2011-08-30 Amgen Inc. Modified Fc molecules
AU2006280321A1 (en) 2005-08-15 2007-02-22 Genentech, Inc. Gene disruptions, compositions and methods relating thereto
CN101297034A (en) 2005-08-24 2008-10-29 洛克菲勒大学 PLY-GBS mutant lysins
CA2624900A1 (en) 2005-10-04 2007-04-19 The Research Foundation Of State University Of New York Fibronectin polypeptides and methods of use
US20080213274A1 (en) * 2005-10-28 2008-09-04 Sabbadini Roger A Compositions and methods for the treatment and prevention of fibrotic, inflammatory, and neovascularization conditions of the eye
MX2008005405A (en) * 2005-10-28 2008-09-11 Florida Internat University Bo Horse: human chimeric antibodies.
US20090074720A1 (en) * 2005-10-28 2009-03-19 Sabbadini Roger A Methods for decreasing immune response and treating immune conditions
RS54111B1 (en) 2005-11-18 2015-12-31 Glenmark Pharmaceuticals S.A. Anti-alpha2 integrin antibodies and their uses
ZA200804162B (en) 2005-11-21 2009-12-30 Genentech Inc Novel gene disruptions, compositions and methods relating thereto
AU2006318539B2 (en) * 2005-11-23 2012-09-13 Genentech, Inc. Methods and compositions related to B cell assays
US20070213264A1 (en) 2005-12-02 2007-09-13 Mingdong Zhou Neuregulin variants and methods of screening and using thereof
ES2664086T3 (en) 2005-12-30 2018-04-18 Zensun (Shanghai) Science & Technology, Co., Ltd. Extended release of neurregulin to improve cardiac function
WO2007102946A2 (en) 2006-01-23 2007-09-13 Amgen Inc. Crystalline polypeptides
WO2007114979A2 (en) 2006-02-17 2007-10-11 Genentech, Inc. Gene disruptons, compositions and methods relating thereto
TW200745163A (en) * 2006-02-17 2007-12-16 Syntonix Pharmaceuticals Inc Peptides that block the binding of IgG to FcRn
TWI417301B (en) 2006-02-21 2013-12-01 Wyeth Corp Antibodies against human il-22 and uses therefor
TW200744634A (en) 2006-02-21 2007-12-16 Wyeth Corp Methods of using antibodies against human IL-22
JP2009529915A (en) 2006-03-20 2009-08-27 ゾーマ テクノロジー リミテッド Human antibodies and methods specific for gastrin substances
TWI397535B (en) 2006-03-21 2013-06-01 Genentech Inc Combinatorial therapy involving alpha5beta1 antagonists
US7973142B2 (en) 2006-04-07 2011-07-05 Warner Chilcott Company Antibodies that bind human protein tyrosine phosphatase beta (HPTPβ)
US20090288176A1 (en) 2006-04-19 2009-11-19 Genentech, Inc. Novel Gene Disruptions, Compositions and Methods Relating Thereto
TWI395754B (en) 2006-04-24 2013-05-11 Amgen Inc Humanized c-kit antibody
US7862812B2 (en) * 2006-05-31 2011-01-04 Lpath, Inc. Methods for decreasing immune response and treating immune conditions
CA2654317A1 (en) 2006-06-12 2007-12-21 Trubion Pharmaceuticals, Inc. Single-chain multivalent binding proteins with effector function
US20080227686A1 (en) * 2006-06-16 2008-09-18 Lipid Sciences, Inc. Novel Peptides that Promote Lipid Efflux
US20080206142A1 (en) * 2006-06-16 2008-08-28 Lipid Sciences, Inc. Novel Peptides That Promote Lipid Efflux
US20080199398A1 (en) * 2006-06-16 2008-08-21 Brewer H Bryan Novel Peptides That Promote Lipid Efflux
US7981425B2 (en) 2006-06-19 2011-07-19 Amgen Inc. Thrombopoietic compounds
CA2787343C (en) 2006-06-26 2016-08-02 Amgen Inc. Compositions comprising modified lcat and uses thereof
EP2057193B1 (en) 2006-08-04 2013-12-18 Novartis AG Ephb3-specific antibody and uses thereof
TWI454480B (en) 2006-08-18 2014-10-01 Novartis Ag Prlr-specific antibody and uses thereof
CL2007002567A1 (en) 2006-09-08 2008-02-01 Amgen Inc ISOLATED PROTEINS FROM LINK TO ACTIVINE TO HUMAN.
WO2008039843A2 (en) * 2006-09-26 2008-04-03 Lipid Sciences, Inc. Novel peptides that promote lipid efflux
BRPI0717512A2 (en) 2006-09-29 2013-11-19 Hoffmann La Roche CCR5 ANTIBODIES AND USES OF THE SAME
US7767206B2 (en) 2006-10-02 2010-08-03 Amgen Inc. Neutralizing determinants of IL-17 Receptor A and antibodies that bind thereto
EP1914303A1 (en) * 2006-10-09 2008-04-23 Qiagen GmbH Thermus eggertssonii DNA polymerases
WO2008055072A2 (en) 2006-10-27 2008-05-08 Lpath, Inc. Compositions and methods for treating ocular diseases and conditions
MX2009004532A (en) 2006-10-27 2009-09-04 Lpath Inc Compositions and methods for binding sphingosine-1-phosphate.
EP3284825B1 (en) 2006-11-02 2021-04-07 Biomolecular Holdings LLC Methods of producing hybrid polypeptides with moving parts
EP2423226A3 (en) 2006-11-10 2012-05-30 Amgen Inc. Antibody-based diagnostics and therapeutics
JP5313909B2 (en) 2006-11-13 2013-10-09 アイコス コーポレイション Thienopyrimidinone for the treatment of inflammatory diseases and cancer
WO2008061019A2 (en) 2006-11-14 2008-05-22 Genentech, Inc. Modulators of neuronal regeneration
EP3156415A1 (en) 2006-11-22 2017-04-19 Bristol-Myers Squibb Company Targeted therapeutics based on engineered proteins for tyrosine kinases receptors, including igf-ir
WO2008070780A1 (en) 2006-12-07 2008-06-12 Novartis Ag Antagonist antibodies against ephb3
US8183201B2 (en) * 2006-12-26 2012-05-22 National Cheng Kung University Methods of treating αvβ3 integrin-associated diseases by administering polypeptides selective for αvβ3 integrin
US7943728B2 (en) * 2006-12-26 2011-05-17 National Cheng Kung University Disintegrin variants and their use in treating osteoporosis-induced bone loss and angiogenesis-related diseases
JP2010517944A (en) * 2007-01-26 2010-05-27 バイオインヴェント インターナショナル アーベー DLL4 signaling inhibitor and use thereof
JP2010517529A (en) * 2007-02-02 2010-05-27 アムジエン・インコーポレーテツド Hepcidin and hepcidin antibody
EP2111228B1 (en) 2007-02-02 2011-07-20 Bristol-Myers Squibb Company 10Fn3 domain for use in treating diseases associated with inappropriate angiogenesis
CN103966345A (en) 2007-02-09 2014-08-06 西北大学 Particles for detecting intracellular targets
US8088887B2 (en) * 2007-02-13 2012-01-03 Academia Sinica Peptide-conjugates that bind to VEGF-stimulated or tumor vasculature and methods of treatment
US8415453B2 (en) * 2007-02-13 2013-04-09 Academia Sinica Lung cancer-targeted peptides and applications thereof
WO2008103962A2 (en) 2007-02-22 2008-08-28 Genentech, Inc. Methods for detecting inflammatory bowel disease
US8808747B2 (en) * 2007-04-17 2014-08-19 Baxter International Inc. Nucleic acid microparticles for pulmonary delivery
CA2689923A1 (en) 2007-05-30 2008-12-11 Northwestern University Nucleic acid functionalized nanoparticles for therapeutic applications
DK2164992T3 (en) * 2007-05-30 2016-08-15 Lpath Inc COMPOSITIONS AND METHODS FOR BONDING OF LYTHOPHOSPHATIC ACID
US9163091B2 (en) * 2007-05-30 2015-10-20 Lpath, Inc. Compositions and methods for binding lysophosphatidic acid
US8759300B2 (en) * 2007-06-14 2014-06-24 The Research Foundation For The State University Of New York Polypeptides and methods of use
US7625555B2 (en) 2007-06-18 2009-12-01 Novagen Holding Corporation Recombinant human interferon-like proteins
EP2167130A2 (en) * 2007-07-06 2010-03-31 Trubion Pharmaceuticals, Inc. Binding peptides having a c-terminally disposed specific binding domain
CN101802013B (en) 2007-07-16 2014-07-02 健泰科生物技术公司 Humanized anti-CD79b antibodies and immunoconjugates and methods of use
NZ583367A (en) 2007-07-16 2012-10-26 Genentech Inc Anti-cd79b antibodies and immunoconjugates and methods of use
PT2181190E (en) 2007-07-26 2014-02-24 Amgen Inc Modified lecithin-cholesterol acyltransferase enzymes
CN101361968B (en) 2007-08-06 2011-08-03 健能隆医药技术(上海)有限公司 Use of interleukin-22 in treating fatty liver
BRPI0814763A2 (en) * 2007-08-09 2015-03-03 Syntonix Pharmaceuticals Inc IMMUNOMODULATING PEPTIDES
EP2615113A3 (en) 2007-08-23 2013-11-13 Amgen Inc. Antigen binding proteins to proprotein convertase subtilisin kexin type 9 (PCSK9)
JOP20080381B1 (en) 2007-08-23 2023-03-28 Amgen Inc Antigen Binding Proteins to Proprotein Convertase subtillisin Kexin type 9 (pcsk9)
NZ583605A (en) 2007-08-29 2012-10-26 Sanofi Aventis Humanized anti-cxcr5 antibodies, derivatives thereof and their uses
US7982016B2 (en) 2007-09-10 2011-07-19 Amgen Inc. Antigen binding proteins capable of binding thymic stromal lymphopoietin
US20090156488A1 (en) 2007-09-12 2009-06-18 Zensun (Shanghai) Science & Technology Limited Use of neuregulin for organ preservation
EP2050764A1 (en) 2007-10-15 2009-04-22 sanofi-aventis Novel polyvalent bispecific antibody format and uses thereof
US8361465B2 (en) * 2007-10-26 2013-01-29 Lpath, Inc. Use of anti-sphingosine-1-phosphate antibodies in combination with chemotherapeutic agents
WO2009058564A2 (en) 2007-11-01 2009-05-07 Maxygen, Inc. Immunosuppressive polypeptides and nucleic acids
US8541543B2 (en) * 2007-11-20 2013-09-24 Academia Sinica Peptides specific for hepatocellular carcinoma cells and applications thereof
EP2231181B1 (en) 2007-12-17 2016-02-17 Marfl AB New vaccine for the treatment of mycobacterium related disorders
WO2009085200A2 (en) 2007-12-21 2009-07-09 Amgen Inc. Anti-amyloid antibodies and uses thereof
SI2808343T1 (en) 2007-12-26 2019-10-30 Xencor Inc Fc variants with altered binding to FcRn
WO2009094551A1 (en) 2008-01-25 2009-07-30 Amgen Inc. Ferroportin antibodies and methods of use
MX2010008437A (en) 2008-01-31 2010-11-25 Genentech Inc Anti-cd79b antibodies and immunoconjugates and methods of use.
JP6018361B2 (en) 2008-01-31 2016-11-02 アンスティチュ ナショナル ドゥ ラ サンテ エ ドゥ ラ ルシェルシュ メディカル Antibodies against human CD39 and their use for inhibiting regulatory T cell activity
JO2913B1 (en) 2008-02-20 2015-09-15 امجين إنك, Antibodies directed to angiopoietin-1 and angiopoietin-2 and uses thereof
US8697081B2 (en) * 2008-04-09 2014-04-15 The Regents Of The University Of Michigan Method of modulating neovascularization
EP2279412B1 (en) 2008-04-09 2017-07-26 Genentech, Inc. Novel compositions and methods for the treatment of immune related diseases
EP2365003A1 (en) * 2008-04-11 2011-09-14 Emergent Product Development Seattle, LLC CD37 immunotherapeutic and combination with bifunctional chemotherapeutic thereof
US8921315B1 (en) 2008-04-24 2014-12-30 Neumedicines, Inc. Method of increasing survival of a human subject having exposure to an acute exposure to non-therapeutic whole body ionization by administering a therapeutically effective dose of IL-12
EP2816059A1 (en) 2008-05-01 2014-12-24 Amgen, Inc Anti-hepcidin antibodies and methods of use
EP3388527A1 (en) 2008-05-15 2018-10-17 Tetherex Pharmaceuticals Corporation Anti-psgl-1 antibodies and methods of identification and use
US8093018B2 (en) 2008-05-20 2012-01-10 Otsuka Pharmaceutical Co., Ltd. Antibody identifying an antigen-bound antibody and an antigen-unbound antibody, and method for preparing the same
JP2011520961A (en) 2008-05-22 2011-07-21 ブリストル−マイヤーズ スクイブ カンパニー Scaffold domain protein based on multivalent fibronectin
JOP20190083A1 (en) 2008-06-04 2017-06-16 Amgen Inc Fgf21 mutant fusion polypeptides and uses thereof
EP2318036B1 (en) 2008-06-30 2015-06-03 The Regents of the University of Michigan Lysosomal phospholipase a2 (lpla2) activity as a diagnostic and therapeutic target for identifying and treating systemic lupus erythematosis
US20100048488A1 (en) * 2008-08-01 2010-02-25 Syntonix Pharmaceuticals, Inc. Immunomodulatory peptides
US8163497B2 (en) 2008-09-07 2012-04-24 Glyconex Inc. Anti-extended type I glycosphingolipid antibody, derivatives thereof and use
SI2344540T1 (en) 2008-10-02 2018-04-30 Aptevo Research And Development Llc Cd86 antagonist multi-target binding proteins
JP2012504946A (en) 2008-10-07 2012-03-01 アンスティチュ ナショナル ドゥ ラ サンテ エ ドゥ ラ ルシェルシュ メディカル Neutralizing antibody against platelet factor 4 variant 1 (PF4V1) and fragments thereof
EA032727B1 (en) 2008-10-10 2019-07-31 Амген Инк. Fgf21 mutant proteolysis-resistant polypeptide and use thereof
AU2009308293B2 (en) 2008-10-22 2015-02-05 Genentech, Inc. Modulation of axon degeneration
US8871202B2 (en) 2008-10-24 2014-10-28 Lpath, Inc. Prevention and treatment of pain using antibodies to sphingosine-1-phosphate
JP5775458B2 (en) * 2008-11-06 2015-09-09 グレンマーク ファーマシューティカルズ, エセ.アー. Treatment using anti-α2 integrin antibody
US20100202963A1 (en) 2008-11-13 2010-08-12 Gallatin W Michael Therapies for hematologic malignancies
US9492449B2 (en) 2008-11-13 2016-11-15 Gilead Calistoga Llc Therapies for hematologic malignancies
KR101692880B1 (en) 2008-11-24 2017-01-04 노오쓰웨스턴 유니버시티 Polyvalent rna-nanoparticle compositions
EP2379096B1 (en) 2008-12-19 2019-10-30 Baxalta GmbH Tfpi inhibitors and methods of use
US20100184844A1 (en) * 2009-01-08 2010-07-22 Northwestern University Inhibition of Bacterial Protein Production by Polyvalent Oligonucleotide Modified Nanoparticle Conjugates
US20100233270A1 (en) 2009-01-08 2010-09-16 Northwestern University Delivery of Oligonucleotide-Functionalized Nanoparticles
US20100294952A1 (en) * 2009-01-15 2010-11-25 Northwestern University Controlled agent release and sequestration
EP3002296B1 (en) 2009-03-17 2020-04-29 Université d'Aix-Marseille Btla antibodies and uses thereof
US20120121591A1 (en) 2009-03-20 2012-05-17 Amgen Inc. SELECTIVE AND POTENT PEPTIDE INHIBITORS OF Kv1.3
KR20120002995A (en) 2009-03-24 2012-01-09 길리아드 칼리스토가 엘엘씨 Atropisomers of 2-purinyl-3-tolyl-quinazolinone derivatives and methods of use
UA108199C2 (en) 2009-03-25 2015-04-10 ANTIBODY AGAINST α5β1 AND ITS APPLICATION
JP5795306B2 (en) 2009-04-01 2015-10-14 ジェネンテック, インコーポレイテッド Treatment of insulin resistance disease
WO2010120561A1 (en) 2009-04-01 2010-10-21 Genentech, Inc. Anti-fcrh5 antibodies and immunoconjugates and methods of use
WO2010112034A2 (en) 2009-04-02 2010-10-07 Aarhus Universitet Compositions and methods for treatment and diagnosis of synucleinopathies
US8067201B2 (en) * 2009-04-17 2011-11-29 Bristol-Myers Squibb Company Methods for protein refolding
CN102458410A (en) * 2009-04-20 2012-05-16 吉联亚·卡利斯托加有限责任公司 Methods of treatment for solid tumors
EP2248903A1 (en) 2009-04-29 2010-11-10 Universitat Autònoma De Barcelona Methods and reagents for efficient and targeted gene transfer to monocytes and macrophages
JP2012525847A (en) 2009-05-05 2012-10-25 アムジエン・インコーポレーテツド FGF21 variants and uses thereof
PE20120358A1 (en) 2009-05-05 2012-04-26 Amgen Inc FGF21 MUTANTS AND USES OF THEM
TW201102086A (en) 2009-06-04 2011-01-16 Hoffmann La Roche Antibodies against human CCN1 and uses thereof
CA2797480A1 (en) 2009-06-15 2010-12-23 4S3 Bioscience Inc. Methods and compositions for treatment of myotubular myopathy using chimeric polypeptides comprising myotubularin 1 (mtm1) polypeptides
CA2764835A1 (en) 2009-06-17 2010-12-23 Amgen Inc. Chimeric fgf19 polypeptides and uses thereof
CN102470156A (en) * 2009-07-20 2012-05-23 成功大学 Polypeptides selective for av ss3 integrin conjugated with a variant of human serum albumin (HSA) and pharmaceutical uses thereof
CA2768843A1 (en) 2009-07-21 2011-01-27 Gilead Calistoga Llc Treatment of liver disorders with pi3k inhibitors
WO2011014750A1 (en) 2009-07-31 2011-02-03 Genentech, Inc. Inhibition of tumor metastasis using bv8- or g-csf-antagonists
WO2011028952A1 (en) 2009-09-02 2011-03-10 Xencor, Inc. Compositions and methods for simultaneous bivalent and monovalent co-engagement of antigens
US8926976B2 (en) 2009-09-25 2015-01-06 Xoma Technology Ltd. Modulators
WO2011038301A2 (en) 2009-09-25 2011-03-31 Xoma Technology Ltd. Screening methods
AU2010308030B2 (en) 2009-10-12 2014-05-29 Pfizer Inc. Cancer treatment
TW201117824A (en) 2009-10-12 2011-06-01 Amgen Inc Use of IL-17 receptor a antigen binding proteins
EP2488643A4 (en) * 2009-10-15 2013-07-03 Hoffmann La Roche Chimeric fibroblast growth factors with altered receptor specificity
WO2011049625A1 (en) 2009-10-20 2011-04-28 Mansour Samadpour Method for aflatoxin screening of products
KR20120105446A (en) 2009-10-22 2012-09-25 제넨테크, 인크. Methods and compositions for modulating hepsin activation of macrophage-stimulating protein
CN104043126A (en) 2009-10-22 2014-09-17 霍夫曼-拉罗奇有限公司 Modulation of axon degeneration
WO2011056502A1 (en) 2009-10-26 2011-05-12 Genentech, Inc. Bone morphogenetic protein receptor type ii compositions and methods of use
WO2011056494A1 (en) 2009-10-26 2011-05-12 Genentech, Inc. Activin receptor-like kinase-1 antagonist and vegfr3 antagonist combinations
WO2011056497A1 (en) 2009-10-26 2011-05-12 Genentech, Inc. Activin receptor type iib compositions and methods of use
KR20120136345A (en) 2009-10-30 2012-12-18 노오쓰웨스턴 유니버시티 Templated nanoconjugates
CA2779574C (en) 2009-11-05 2018-12-18 Rhizen Pharmaceuticals S.A. Novel kinase modulators
RU2012124093A (en) 2009-11-12 2013-12-20 Дженентек, Инк. METHOD FOR INCREASING DENSITY OF DENDRITIC SPIKES
US9260517B2 (en) 2009-11-17 2016-02-16 Musc Foundation For Research Development Human monoclonal antibodies to human nucleolin
TW201129379A (en) 2009-11-20 2011-09-01 Amgen Inc Anti-Orai1 antigen binding proteins and uses thereof
AU2010324686B2 (en) 2009-11-30 2016-05-19 Genentech, Inc. Antibodies for treating and diagnosing tumors expressing SLC34A2 (TAT211 = SEQID2 )
WO2011066511A1 (en) 2009-11-30 2011-06-03 The U.S.A., As Represented By The Secretary Department Of Health And Human Services Synthetic apoa-1 mimetic amphipathic peptides and methods of use thereof
EP2506861A1 (en) * 2009-12-02 2012-10-10 Amgen Inc. Binding proteins that bind to human fgfr1c, human b-klotho and both human fgfr1c and human b-klotho
UA109888C2 (en) 2009-12-07 2015-10-26 ANTIBODY OR ANTIBODILITY ANTIBODY OR ITS BINDING TO THE β-CLOTE, FGF RECEPTORS AND THEIR COMPLEXES
AU2010330794A1 (en) 2009-12-18 2012-06-21 Amgen Inc. Wise binding agents and epitopes
HUE029026T2 (en) 2009-12-22 2017-01-30 Roche Glycart Ag ANTI-HER3 Antibodies and uses thereof
MX2012007416A (en) 2009-12-23 2012-07-23 Univ Nat Cheng Kung Compositions and methods for the treatment of angiogenesis-related eye diseases.
WO2011079308A2 (en) 2009-12-23 2011-06-30 Emergent Product Development Seattle, Llc Compositions comprising tnf-alpha and il-6 antagonists and methods of use thereof
JP5856073B2 (en) 2009-12-29 2016-02-09 エマージェント プロダクト デベロップメント シアトル, エルエルシー RON binding construct and method of use thereof
WO2011097527A2 (en) 2010-02-04 2011-08-11 Xencor, Inc. Immunoprotection of therapeutic moieties using enhanced fc regions
US20110189178A1 (en) * 2010-02-04 2011-08-04 Xencor, Inc. Immunoprotection of Therapeutic Moieties Using Enhanced Fc Regions
CN102781960B (en) 2010-02-16 2014-12-10 米迪缪尼有限公司 HSA-related compositions and methods of use
US8877897B2 (en) 2010-02-23 2014-11-04 Genentech, Inc. Compositions and methods for the diagnosis and treatment of tumor
US8975376B2 (en) 2010-02-23 2015-03-10 Sanofi Anti-alpha2 integrin antibodies and their uses
US20110212088A1 (en) * 2010-02-26 2011-09-01 Sabbadini Roger A Anti-paf antibodies
US8642557B2 (en) 2010-03-12 2014-02-04 Abbvie Biotherapeutics Inc. CTLA4 proteins and their uses
CN103025345B (en) 2010-03-19 2016-01-20 巴克斯特国际公司 TFPI inhibitor and using method
MY187990A (en) 2010-03-31 2021-11-07 Boehringer Ingelheim Int Anti-cd40 antibodies
JP2013523726A (en) 2010-04-01 2013-06-17 オンコレナ エービー Improved treatment of renal cell carcinoma
US9517264B2 (en) 2010-04-15 2016-12-13 Amgen Inc. Human FGF receptor and β-Klotho binding proteins
BR112012028010A2 (en) 2010-05-03 2017-09-26 Genentech Inc isolated antibody, cell, isolated nucleic acid, method of identifying a first antibody that binds to a tat425 antigenic epitope attached to an antibody, methods of inhibiting cell growth, therapeutic treatment of determining the presence of a tat425 protein and diagnosing the presence of a tumor in a mammal
EP2571878B1 (en) 2010-05-17 2018-10-17 Incozen Therapeutics Pvt. Ltd. Novel 3,5-disubstitued-3h-imidazo[4,5-b]pyridine and 3,5- disubstitued -3h-[1,2,3]triazolo[4,5-b]pyridine compounds as modulators of protein kinases
DK2571516T3 (en) 2010-05-18 2018-02-05 Neumedicines Inc IL-12 FORMULATIONS FOR STIMULATING HEMOPOIES
EP2576615B1 (en) 2010-05-26 2016-03-30 Bristol-Myers Squibb Company Fibronectin based scaffold proteins having improved stability
WO2011160062A2 (en) 2010-06-17 2011-12-22 The Usa As Represented By The Secretary, National Institutes Of Health Compositions and methods for treating inflammatory conditions
AR082163A1 (en) 2010-07-15 2012-11-14 Hoffmann La Roche SPECIFICALLY BINDING ANTIBODIES OF THE HUMAN TSLPR AND METHODS OF USING THEMSELVES
WO2012010696A1 (en) 2010-07-23 2012-01-26 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods for cancer management targeting co-029
CA2806252C (en) 2010-07-29 2019-05-14 Xencor, Inc. Antibodies with modified isoelectric points
US20130177555A1 (en) 2010-08-13 2013-07-11 Medimmune Limited Monomeric Polypeptides Comprising Variant FC Regions And Methods Of Use
US9688735B2 (en) 2010-08-20 2017-06-27 Wyeth Llc Designer osteogenic proteins
KR101630501B1 (en) 2010-08-20 2016-06-15 와이어쓰 엘엘씨 Designer osteogenic proteins
CN102380091A (en) 2010-08-31 2012-03-21 健能隆医药技术(上海)有限公司 Application of interleukin-22 in curing virus hepatitis
JP6159660B2 (en) 2010-09-22 2017-07-05 アムジエン・インコーポレーテツド Immunoglobulins as carriers and uses thereof
EP2621954A1 (en) 2010-10-01 2013-08-07 Oxford Biotherapeutics Ltd. Anti-rori antibodies
US9445990B2 (en) 2010-10-06 2016-09-20 Medtronic, Inc. TNF inhibitor formulation for use in implantable infusion devices
WO2012061129A1 (en) 2010-10-25 2012-05-10 Genentech, Inc Treatment of gastrointestinal inflammation and psoriasis a
EA201890548A1 (en) 2010-11-04 2018-07-31 Бёрингер Ингельхайм Интернациональ Гмбх ANTIBODIES TO IL-23
US9023791B2 (en) 2010-11-19 2015-05-05 Novartis Ag Fibroblast growth factor 21 mutations
WO2012080769A1 (en) 2010-12-15 2012-06-21 INSERM (Institut National de la Santé et de la Recherche Médicale) Anti-cd277 antibodies and uses thereof
CN108939067A (en) 2010-12-21 2018-12-07 瑟莱克斯制药公司 Anti- palatelet-selectin antibody and its use and identification method
WO2012085132A1 (en) 2010-12-22 2012-06-28 Orega Biotech Antibodies against human cd39 and use thereof
JOP20210044A1 (en) 2010-12-30 2017-06-16 Takeda Pharmaceuticals Co Anti-cd38 antibodies
WO2012101125A1 (en) 2011-01-24 2012-08-02 INSERM (Institut National de la Santé et de la Recherche Médicale) Specific antibodies against human cxcl4 and uses thereof
WO2012102679A1 (en) 2011-01-24 2012-08-02 National University Of Singapore Pathogenic mycobacteria-derived mannose-capped lipoarabinomannan antigen binding proteins
TW201238976A (en) 2011-02-23 2012-10-01 Hoffmann La Roche Antibodies against human IL33R and uses thereof
AR085911A1 (en) 2011-03-16 2013-11-06 Sanofi Sa SAFE THERAPEUTIC DOSE OF A SIMILAR PROTEIN TO AN ANTIBODY WITH VUAL REGION
EA035351B1 (en) 2011-03-31 2020-06-01 Инсэрм (Инститют Насиональ Де Ля Сантэ Э Де Ля Решерш Медикаль) Antibodies directed against icos and uses thereof
KR102001686B1 (en) 2011-04-07 2019-07-18 암젠 인크 Novel egfr binding proteins
TR201905909T4 (en) 2011-04-19 2019-05-21 Pfizer Combinations of anti-4-1bb antibodies and adcc inducing antibodies for cancer therapy.
MX365160B (en) 2011-05-04 2019-05-24 Rhizen Pharmaceuticals Sa Novel compounds as modulators of protein kinases.
JOP20200043A1 (en) 2011-05-10 2017-06-16 Amgen Inc Methods of treating or preventing cholesterol related disorders
MX344219B (en) 2011-05-18 2016-12-07 Mederis Diabetes Llc Improved peptide pharmaceuticals for insulin resistance.
EP4286400A2 (en) 2011-05-18 2023-12-06 Eumederis Pharmaceuticals, Inc. Improved peptide pharmaceuticals
US9127065B2 (en) 2011-05-19 2015-09-08 Institut National De La Sante Et De La Recherche Medicale (Inserm) Anti-human HER3 antibodies and uses thereof
ES2894398T3 (en) 2011-06-03 2022-02-14 Xoma Technology Ltd Specific antibodies to TGF-beta
WO2012171031A1 (en) 2011-06-10 2012-12-13 Baxter International Inc. Treatment of coagulation disease by administration of recombinant vwf
EP2718456B1 (en) 2011-06-13 2019-04-24 Neumedicines, Inc. Mitigation of cutaneous injury with il-12
US9045526B2 (en) 2011-06-23 2015-06-02 The Regents Of The University Of Michigan Compound and method for modulating opioid receptor activity
EP2543678A1 (en) 2011-07-08 2013-01-09 INSERM (Institut National de la Santé et de la Recherche Médicale) Antibodies for the treatment and prevention of thrombosis
EP2543677A1 (en) 2011-07-08 2013-01-09 INSERM (Institut National de la Santé et de la Recherche Médicale) Antibodies for the treatment and prevention of thrombosis
EP2543679A1 (en) 2011-07-08 2013-01-09 INSERM (Institut National de la Santé et de la Recherche Médicale) Antibodies for the treatment and prevention of thrombosis
WO2013012855A1 (en) 2011-07-18 2013-01-24 Amgen Inc. Apelin antigen-binding proteins and uses thereof
US20140234330A1 (en) 2011-07-22 2014-08-21 Amgen Inc. Il-17 receptor a is required for il-17c biology
WO2013022855A1 (en) 2011-08-05 2013-02-14 Xencor, Inc. Antibodies with modified isoelectric points and immunofiltering
US9605083B2 (en) 2011-08-16 2017-03-28 Emory University JAML specific binding agents, antibodies, and uses related thereto
KR20140068877A (en) 2011-08-17 2014-06-09 제넨테크, 인크. Inhibition of angiogenesis in refractory tumors
AU2012301769B2 (en) 2011-08-31 2016-05-19 Amgen Inc. FGF21 for use in treating type 1 diabetes
AU2012308302A1 (en) 2011-09-14 2014-03-20 Northwestern University Nanoconjugates able to cross the blood-brain barrier
EA027900B1 (en) 2011-09-22 2017-09-29 Эмджен Инк. Cd27l antigen binding proteins
US9458214B2 (en) 2011-09-26 2016-10-04 Novartis Ag Dual function fibroblast growth factor 21 proteins
US10851178B2 (en) 2011-10-10 2020-12-01 Xencor, Inc. Heterodimeric human IgG1 polypeptides with isoelectric point modifications
WO2013053076A1 (en) 2011-10-10 2013-04-18 Zensun (Shanghai)Science & Technology Limited Compositions and methods for treating heart failure
AU2012323287B2 (en) 2011-10-10 2018-02-01 Xencor, Inc. A method for purifying antibodies
US8999325B2 (en) 2011-10-13 2015-04-07 Aerpio Therapeutics, Inc Treatment of ocular disease
CN104039351A (en) 2011-10-13 2014-09-10 阿尔皮奥治疗学股份有限公司 Methods for treating vascular leak syndrome and cancer
CN104053670A (en) 2011-10-31 2014-09-17 百时美施贵宝公司 Fibronectin binding domains with reduced immunogenicity
SG11201402283PA (en) 2011-11-16 2014-06-27 Boehringer Ingelheim Int Anti il-36r antibodies
MX2014006272A (en) 2011-11-23 2014-10-24 Igenica Biotherapeutics Inc Anti-cd98 antibodies and methods of use thereof.
JP2015502368A (en) 2011-12-16 2015-01-22 カロス セラピューティクス,インコーポレーテッド Methods and uses of ANP (atrial natriuretic peptide), BNP (brain natriuretic peptide) and CNP (C-type natriuretic peptide) -related peptides and their derivatives for the treatment of retinal disorders and diseases
US9988439B2 (en) 2011-12-23 2018-06-05 Nicholas B. Lydon Immunoglobulins and variants directed against pathogenic microbes
EP2793944A4 (en) 2011-12-23 2015-09-02 Nicholas B Lydon Immunoglobulins and variants directed against pathogenic microbes
US9636381B2 (en) 2012-01-18 2017-05-02 Neumedicines, Inc. Methods for radiation protection by administering IL-12
EP2807187B1 (en) 2012-01-26 2017-07-26 Christopher J. Soares Peptide antagonists of the calcitonin cgrp family of peptide hormones and their use
SI2834241T1 (en) 2012-03-05 2021-06-30 Gilead Calistoga Llc Polymorphic forms of (s)-2-(1-(9h-purin-6-ylamino)propyl)-5-fluoro-3-phenylquinazolin-4(3h)-one
KR102143887B1 (en) 2012-03-16 2020-08-12 유니버시티 헬스 네트워크 Methods and compositions for modulating toso activity
CN104302306B (en) 2012-03-21 2019-03-22 百深有限责任公司 TFPI inhibitor and its application method
US9592289B2 (en) 2012-03-26 2017-03-14 Sanofi Stable IgG4 based binding agent formulations
CA2868883C (en) 2012-03-30 2022-10-04 Sorrento Therapeutics Inc. Fully human antibodies that bind to vegfr2
KR20140144726A (en) 2012-03-30 2014-12-19 리젠 파마슈티컬스 소시에떼 아노님 Novel 3,5-disubstitued-3h-imidazo[4,5-b]pyridine and 3,5-disubstitued -3h-[1,2,3]triazolo[4,5-b] pyridine compounds as modulators of c-met protein kinases
WO2013151649A1 (en) 2012-04-04 2013-10-10 Sialix Inc Glycan-interacting compounds
WO2013155346A1 (en) 2012-04-11 2013-10-17 The Regents Of The University Of California Diagnostic tools for response to 6-thiopurine therapy
EA039663B1 (en) 2012-05-03 2022-02-24 Амген Инк. Use of an anti-pcsk9 antibody for lowering serum cholesterol ldl and treating cholesterol related disorders
CN109206516A (en) 2012-05-03 2019-01-15 勃林格殷格翰国际有限公司 Anti-il-23 p 19 antibodies
EP2847219A1 (en) 2012-05-07 2015-03-18 Amgen Inc. Anti-erythropoietin antibodies
EP2850095B1 (en) 2012-05-17 2019-10-09 RA Pharmaceuticals, Inc. Peptide and peptidomimetic inhibitors
UY34813A (en) 2012-05-18 2013-11-29 Amgen Inc ANTIGEN UNION PROTEINS DIRECTED AGAINST ST2 RECEIVER
EP3553086A1 (en) 2012-05-31 2019-10-16 Sorrento Therapeutics Inc. Antigen binding proteins that bind pd-l1
KR20150030706A (en) 2012-06-11 2015-03-20 암젠 인코퍼레이티드 Dual receptor antagonistic antigen-binding proteins and uses thereof
CA2877573A1 (en) 2012-06-21 2013-12-27 Sorrento Therapeutics, Inc. Antigen binding proteins that bind c-met
US9315579B2 (en) 2012-06-22 2016-04-19 Sorrento Therapeutics, Inc. Antigen binding proteins that bind CCR2
EP2864355B1 (en) 2012-06-25 2016-10-12 Orega Biotech Il-17 antagonist antibodies
WO2014004549A2 (en) 2012-06-27 2014-01-03 Amgen Inc. Anti-mesothelin binding proteins
EP3431497B1 (en) 2012-06-27 2022-07-27 The Trustees of Princeton University Split inteins, conjugates and uses thereof
WO2014039189A1 (en) 2012-08-01 2014-03-13 Mcnally Elizabeth Mitigating tissue damage and fibrosis via latent transforming growth factor beta binding protein (ltbp4)
WO2014022759A1 (en) 2012-08-03 2014-02-06 Dana-Farber Cancer Institute, Inc. Agents that modulate immune cell activation and methods of use thereof
EP2892928B1 (en) 2012-09-03 2018-05-30 INSERM - Institut National de la Santé et de la Recherche Médicale Antibodies directed against icos for treating graft-versus-host disease
TWI595007B (en) 2012-09-10 2017-08-11 Neotope Biosciences Ltd Anti-mcam antibodies and associated methods of use
EP3738605A1 (en) 2012-09-10 2020-11-18 Xencor, Inc. Methods of treating neurological diseases
EP2906598A1 (en) 2012-10-09 2015-08-19 Igenica Biotherapeutics, Inc. Anti-c16orf54 antibodies and methods of use thereof
MX360816B (en) 2012-11-20 2018-11-15 Mederis Diabetes Llc Improved peptide pharmaceuticals for insulin resistance.
EP2922877B1 (en) 2012-11-20 2018-09-05 Eumederis Pharmaceuticals, Inc. Improved peptide pharmaceuticals
JP6324399B2 (en) 2012-11-20 2018-05-16 サノフイ Anti-CEACAM5 antibody and use thereof
TW201425336A (en) 2012-12-07 2014-07-01 Amgen Inc BCMA antigen binding proteins
EA201591219A1 (en) 2012-12-27 2015-12-30 Санофи ANTIBODIES AGAINST LAMP1 AND CONJUGATES ANTIBODIES AND MEDICINES AND THEIR APPLICATION
US9701759B2 (en) 2013-01-14 2017-07-11 Xencor, Inc. Heterodimeric proteins
US11053316B2 (en) 2013-01-14 2021-07-06 Xencor, Inc. Optimized antibody variable regions
US10131710B2 (en) 2013-01-14 2018-11-20 Xencor, Inc. Optimized antibody variable regions
US10487155B2 (en) 2013-01-14 2019-11-26 Xencor, Inc. Heterodimeric proteins
US10968276B2 (en) 2013-03-12 2021-04-06 Xencor, Inc. Optimized anti-CD3 variable regions
US9605084B2 (en) 2013-03-15 2017-03-28 Xencor, Inc. Heterodimeric proteins
AU2014205086B2 (en) 2013-01-14 2019-04-18 Xencor, Inc. Novel heterodimeric proteins
CA2897987A1 (en) 2013-01-15 2014-07-24 Xencor, Inc. Rapid clearance of antigen complexes using novel antibodies
JO3519B1 (en) 2013-01-25 2020-07-05 Amgen Inc Antibody constructs for CDH19 and CD3
ES2728936T3 (en) 2013-01-25 2019-10-29 Amgen Inc Antibodies directed against CDH19 for melanoma
CA2899889A1 (en) 2013-02-01 2014-08-07 Santa Maria Biotherapeutics, Inc. Administration of an anti-activin-a compound to a subject
US20150361159A1 (en) 2013-02-01 2015-12-17 Bristol-Myers Squibb Company Fibronectin based scaffold proteins
US9580486B2 (en) 2013-03-14 2017-02-28 Amgen Inc. Interleukin-2 muteins for the expansion of T-regulatory cells
ES2759061T3 (en) 2013-03-15 2020-05-07 Biomolecular Holdings Llc Hybrid immunoglobulin containing non-peptidyl binding
SI2970449T1 (en) 2013-03-15 2019-11-29 Amgen Res Munich Gmbh Single chain binding molecules comprising n-terminal abp
UA118843C2 (en) 2013-03-15 2019-03-25 Дженентек, Інк. Il-22 polypeptides and il-22 fc fusion proteins and methods of use
EP2970486B1 (en) 2013-03-15 2018-05-16 Xencor, Inc. Modulation of t cells with bispecific antibodies and fc fusions
US10519242B2 (en) 2013-03-15 2019-12-31 Xencor, Inc. Targeting regulatory T cells with heterodimeric proteins
WO2014152006A2 (en) 2013-03-15 2014-09-25 Intrinsic Lifesciences, Llc Anti-hepcidin antibodies and uses thereof
US10106624B2 (en) 2013-03-15 2018-10-23 Xencor, Inc. Heterodimeric proteins
EP2970446A1 (en) 2013-03-15 2016-01-20 Amgen Research (Munich) GmbH Antibody constructs for influenza m2 and cd3
PL3587448T3 (en) 2013-03-15 2021-11-29 Xencor, Inc. Heterodimeric proteins
US9260527B2 (en) 2013-03-15 2016-02-16 Sdix, Llc Anti-human CXCR4 antibodies and methods of making same
US10858417B2 (en) 2013-03-15 2020-12-08 Xencor, Inc. Heterodimeric proteins
SG10201800800YA (en) 2013-05-06 2018-03-28 Scholar Rock Inc Compositions and methods for growth factor modulation
US10005839B2 (en) 2013-05-17 2018-06-26 Inserm (Institut National De La Sante Et De La Recherche Medicale) Antagonist of the BTLA/HVEM interaction for use in therapy
PL3004167T3 (en) 2013-05-30 2019-01-31 Kiniksa Pharmaceuticals, Ltd. Oncostatin m receptor antigen binding proteins
CN111423511B (en) 2013-05-31 2024-02-23 索伦托药业有限公司 Antigen binding proteins that bind to PD-1
JP6267792B2 (en) 2013-06-28 2018-01-24 アムジエン・インコーポレーテツド Methods for treating homozygous familial hypercholesterolemia
AR097648A1 (en) 2013-09-13 2016-04-06 Amgen Inc COMBINATION OF EPIGENETIC FACTORS AND BIESPECTIVE COMPOUNDS THAT HAVE LIKE DIANA CD33 AND CD3 IN THE TREATMENT OF MYELOID LEUKEMIA
EP3757130A1 (en) 2013-09-26 2020-12-30 Costim Pharmaceuticals Inc. Methods for treating hematologic cancers
WO2015049355A1 (en) 2013-10-04 2015-04-09 Roche Diagnostics Gmbh Antibodies specifically binding to her3
DK3055331T3 (en) 2013-10-11 2021-03-22 Oxford Bio Therapeutics Ltd CONJUGATED ANTIBODIES TO LY75 FOR CANCER TREATMENT
WO2015057583A1 (en) 2013-10-14 2015-04-23 The United States Of America, As Represented By The Secretary Treatment of chronic kidney disease with sahps
WO2015057908A1 (en) 2013-10-18 2015-04-23 Novartis Ag Methods of treating diabetes and related disorders
EP3063317B1 (en) 2013-10-28 2020-06-03 DOTS Technology Corp. Allergen detection
WO2015066550A1 (en) 2013-10-31 2015-05-07 Resolve Therapeutics, Llc Therapeutic nuclease-albumin fusions and methods
CN104623637A (en) 2013-11-07 2015-05-20 健能隆医药技术(上海)有限公司 Application of IL-22 dimer in preparation of intravenous injection drugs
EP3083623A1 (en) 2013-12-20 2016-10-26 Gilead Calistoga LLC Polymorphic forms of a hydrochloride salt of (s) -2-(9h-purin-6-ylamino) propyl) -5-fluoro-3-phenylquinazolin-4 (3h) -one
NZ720867A (en) 2013-12-20 2018-01-26 Gilead Calistoga Llc Process methods for phosphatidylinositol 3-kinase inhibitors
AU2015204446A1 (en) 2014-01-13 2016-07-14 Valerion Therapeutics, Llc Internalizing moieties
RU2701434C2 (en) 2014-01-24 2019-09-26 Нгм Биофармасьютикалс, Инк. Binding proteins and methods for use thereof
CN105873949A (en) 2014-01-31 2016-08-17 勃林格殷格翰国际有限公司 Novel anti-BAFF antibodies
EP4008726A1 (en) 2014-02-20 2022-06-08 Allergan, Inc. Complement component c5 antibodies
RU2018135371A (en) 2014-02-27 2018-12-10 Аллерган, Инк. ANTIBODIES TO THE COMPLEX FACTOR Bb
KR20160127817A (en) 2014-03-07 2016-11-04 유니버시티 헬스 네트워크 Methods and compositions for modifying the immune response
AU2015229186B2 (en) 2014-03-14 2021-01-28 Biomolecular Holdings Llc Hybrid immunoglobulin containing non-peptidyl linkage
RS59907B1 (en) 2014-03-28 2020-03-31 Xencor Inc Bispecific antibodies that bind to cd38 and cd3
US10544231B2 (en) 2014-04-16 2020-01-28 INSERM (Institut National de la Santé et de la Recherche Médicale) Antibodies for the prevention or the treatment of bleeding episodes
FR3020063A1 (en) 2014-04-16 2015-10-23 Gamamabs Pharma ANTI-HER4 HUMAN ANTIBODY
US9856306B2 (en) 2014-05-28 2018-01-02 Spitfire Pharma, Inc. Peptide pharmaceuticals for insulin resistance
CN106714830B (en) 2014-05-30 2020-08-25 上海复宏汉霖生物技术股份有限公司 anti-Epidermal Growth Factor Receptor (EGFR) antibodies
US10106579B2 (en) 2014-06-12 2018-10-23 Ra Pharmaceuticals, Inc. Modulation of complement activity
US11021467B2 (en) 2014-06-13 2021-06-01 Gilead Sciences, Inc. Phosphatidylinositol 3-kinase inhibitors
JP6655074B2 (en) 2014-06-20 2020-02-26 ジェネンテック, インコーポレイテッド Scugacin-based scaffold compositions, methods and uses
AR101669A1 (en) 2014-07-31 2017-01-04 Amgen Res (Munich) Gmbh ANTIBODY CONSTRUCTS FOR CDH19 AND CD3
TW201609811A (en) 2014-07-31 2016-03-16 安美基研究(慕尼黑)公司 Bispecific single chain antibody construct with enhanced tissue distribution
MX2017001401A (en) 2014-07-31 2017-08-07 Amgen Res (Munich) Gmbh Optimized cross-species specific bispecific single chain antibody constructs.
AU2015305894A1 (en) 2014-08-22 2017-04-06 Sorrento Therapeutics, Inc. Antigen binding proteins that bind CXCR3
AU2015306608B2 (en) 2014-08-27 2020-03-05 Amgen Inc. Variants of tissue inhibitor of metalloproteinase type three (TIMP-3), compositions and methods
US10323088B2 (en) 2014-09-22 2019-06-18 Intrinsic Lifesciences Llc Humanized anti-hepcidin antibodies and uses thereof
US20190194654A1 (en) 2014-10-24 2019-06-27 Astrazeneca Ab Combination
CA2967595A1 (en) 2014-11-12 2016-05-19 Siamab Therapeutics, Inc. Glycan-interacting compounds and methods of use
US9879087B2 (en) 2014-11-12 2018-01-30 Siamab Therapeutics, Inc. Glycan-interacting compounds and methods of use
AU2015349680A1 (en) 2014-11-21 2017-06-08 Northwestern University The sequence-specific cellular uptake of spherical nucleic acid nanoparticle conjugates
CN116333153A (en) 2014-11-26 2023-06-27 森科股份有限公司 Heterodimeric antibodies that bind CD3 and tumor antigens
US10259887B2 (en) 2014-11-26 2019-04-16 Xencor, Inc. Heterodimeric antibodies that bind CD3 and tumor antigens
KR20170084327A (en) 2014-11-26 2017-07-19 젠코어 인코포레이티드 Heterodimeric antibodies that bind cd3 and cd38
CN105669863B (en) 2014-12-05 2019-09-13 鸿运华宁(杭州)生物医药有限公司 It is a kind of can with human endothelin receptor specifically bind antibody and its application
US11220545B2 (en) 2014-12-08 2022-01-11 Dana-Farber Cancer Institute, Inc. Methods for upregulating immune responses using combinations of anti-RGMb and anti-PD-1 agents
US11697825B2 (en) 2014-12-12 2023-07-11 Voyager Therapeutics, Inc. Compositions and methods for the production of scAAV
US10428155B2 (en) 2014-12-22 2019-10-01 Xencor, Inc. Trispecific antibodies
EP3916017A1 (en) 2014-12-22 2021-12-01 PD-1 Acquisition Group, LLC Anti-pd-1 antibodies
EP3240801B1 (en) 2014-12-31 2021-01-20 Checkmate Pharmaceuticals, Inc. Combination tumor immunotherapy
PL3247725T3 (en) 2015-01-23 2021-01-11 Sanofi Anti-cd3 antibodies, anti-cd123 antibodies and bispecific antibodies specifically binding to cd3 and/or cd123
US9937222B2 (en) 2015-01-28 2018-04-10 Ra Pharmaceuticals, Inc. Modulators of complement activity
WO2016128349A1 (en) 2015-02-09 2016-08-18 INSERM (Institut National de la Santé et de la Recherche Médicale) Antibodies specific to glycoprotein (gp) of ebolavirus and uses for the treatment and diagnosis of ebola virus infection
AR103675A1 (en) 2015-02-13 2017-05-24 Sorrento Therapeutics Inc ANTI-CTLA4 THERAPEUTIC ANTIBODIES
WO2016138160A1 (en) 2015-02-24 2016-09-01 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Middle east respiratory syndrome coronavirus immunogens, antibodies, and their use
WO2016141387A1 (en) 2015-03-05 2016-09-09 Xencor, Inc. Modulation of t cells with bispecific antibodies and fc fusions
JP7068825B2 (en) 2015-04-08 2022-05-17 ソレント・セラピューティクス・インコーポレイテッド Antibodies therapeutic agents that bind to CD38
CR20170510A (en) 2015-04-10 2018-02-26 Amgen Inc INTERUQUINE MUTEINS 2 FOR THE EXPANSION OF REGULATORY T-CELLS
WO2016164937A2 (en) 2015-04-10 2016-10-13 Amgen Inc. Interleukin-2 muteins for the expansion of t-regulatory cells
DK3283524T3 (en) 2015-04-17 2023-05-30 Amgen Res Munich Gmbh BISPECIFIC ANTIBODY CONSTRUCTIONS AGAINST CDH3 and CD3
US20160347848A1 (en) 2015-05-28 2016-12-01 Medimmune Limited Therapeutic combinations and methods for treating neoplasia
CA2988588A1 (en) 2015-06-12 2016-12-15 Georgia State University Research Foundation, Inc. Compositions and methods for treating opioid tolerance
ES2828694T3 (en) 2015-07-29 2021-05-27 Allergan Inc Heavy chain-only antibodies to ANG-2
TWI796283B (en) 2015-07-31 2023-03-21 德商安美基研究(慕尼黑)公司 Antibody constructs for msln and cd3
TW202346349A (en) 2015-07-31 2023-12-01 德商安美基研究(慕尼黑)公司 Antibody constructs for dll3 and cd3
TWI829617B (en) 2015-07-31 2024-01-21 德商安美基研究(慕尼黑)公司 Antibody constructs for flt3 and cd3
TWI717375B (en) 2015-07-31 2021-02-01 德商安美基研究(慕尼黑)公司 Antibody constructs for cd70 and cd3
TWI744242B (en) 2015-07-31 2021-11-01 德商安美基研究(慕尼黑)公司 Antibody constructs for egfrviii and cd3
US10308711B2 (en) 2015-08-14 2019-06-04 Allergan, Inc. Heavy chain only antibodies to PDGF
WO2017040566A1 (en) 2015-09-01 2017-03-09 Boehringer Ingelheim International Gmbh Use of anti-cd40 antibodies for treatment of lupus nephritis
TWI799366B (en) 2015-09-15 2023-04-21 美商建南德克公司 Cystine knot scaffold platform
EA201890613A1 (en) 2015-09-21 2018-10-31 Аптево Рисёрч Энд Девелопмент Ллс POLYPEPTIDES CONNECTING CD3
JP2018535655A (en) 2015-09-29 2018-12-06 アムジエン・インコーポレーテツド ASGR inhibitor
AU2016332725A1 (en) 2015-09-29 2018-03-22 Celgene Corporation PD-1 binding proteins and methods of use thereof
EP3355908A1 (en) 2015-10-01 2018-08-08 Amgen Inc. Treatment of bile acid disorders
EP3359575B1 (en) 2015-10-09 2020-07-22 Florida State University Research Foundation, Inc. Antibodies specific for 4,6-diamino-5-(formylamino) pyrimidine and uses thereof
EP3362074B1 (en) 2015-10-16 2023-08-09 President and Fellows of Harvard College Regulatory t cell pd-1 modulation for regulating t cell effector immune responses
KR20180088381A (en) 2015-11-12 2018-08-03 시아맙 쎄라퓨틱스, 인코포레이티드 Glycan-interacting compounds and methods of use
KR20180093010A (en) 2015-12-04 2018-08-20 더 리전트 오브 더 유니버시티 오브 캘리포니아 New Cancer Therapeutic Antibodies
US11623957B2 (en) 2015-12-07 2023-04-11 Xencor, Inc. Heterodimeric antibodies that bind CD3 and PSMA
TWI745320B (en) 2015-12-16 2021-11-11 美商Ra製藥公司 Modulators of complement activity
CN116333124A (en) 2016-01-29 2023-06-27 索伦托药业有限公司 Antigen binding proteins that bind to PD-L1
MX2018009386A (en) 2016-02-03 2018-11-09 Amgen Res Munich Gmbh Psma and cd3 bispecific t cell engaging antibody constructs.
KR20180103084A (en) 2016-02-03 2018-09-18 암젠 리서치 (뮌헨) 게엠베하 BCMA and CD3 bispecific T cell engrafting antibody constructs
EA039859B1 (en) 2016-02-03 2022-03-21 Эмджен Рисерч (Мюник) Гмбх Bispecific antibody constructs binding egfrviii and cd3
JP7157981B2 (en) 2016-03-07 2022-10-21 チャールストンファーマ, エルエルシー anti-nucleolin antibody
CN109328069B (en) 2016-04-15 2023-09-01 亿一生物医药开发(上海)有限公司 Use of IL-22 in the treatment of necrotizing enterocolitis
JOP20170091B1 (en) 2016-04-19 2021-08-17 Amgen Res Munich Gmbh Administration of a bispecific construct binding to CD33 and CD3 for use in a method for the treatment of myeloid leukemia
TWI826351B (en) 2016-05-31 2023-12-21 大陸商鴻運華寧(杭州)生物醫藥有限公司 R antibodies, their pharmaceutical compositions and uses
RU2767357C2 (en) 2016-06-14 2022-03-17 Ксенкор, Инк. Bispecific checkpoint inhibitors antibodies
EP3475304B1 (en) 2016-06-28 2022-03-23 Xencor, Inc. Heterodimeric antibodies that bind somatostatin receptor 2
MA45674A (en) 2016-07-15 2019-05-22 Takeda Pharmaceuticals Co METHODS AND MATERIALS FOR EVALUATING A RESPONSE TO PLASMOBLAST AND PLASMOCYTE DEPLÉTION TREATMENTS
TWI790206B (en) 2016-07-18 2023-01-21 法商賽諾菲公司 Bispecific antibody-like binding proteins specifically binding to cd3 and cd123
US10793632B2 (en) 2016-08-30 2020-10-06 Xencor, Inc. Bispecific immunomodulatory antibodies that bind costimulatory and checkpoint receptors
ES2965087T3 (en) 2016-09-02 2024-04-10 Soares Christopher J Use of CGRP receptor antagonists in the treatment of glaucoma
JP2019534858A (en) 2016-09-09 2019-12-05 ジェネンテック, インコーポレイテッド Selective peptide inhibitor of FRIZZLED
AU2017327828B2 (en) 2016-09-16 2023-11-16 Shanghai Henlius Biotech, Inc. Anti-PD-1 antibodies
WO2018053405A1 (en) 2016-09-19 2018-03-22 Celgene Corporation Methods of treating immune disorders using pd-1 binding proteins
JP2019534859A (en) 2016-09-19 2019-12-05 セルジーン コーポレイション Method for treating vitiligo using PD-1 binding protein
JP2019537621A (en) 2016-10-04 2019-12-26 フェアバンクス ファーマシューティカルズ,インコーポレイテッド Anti-FSTL3 antibodies and uses thereof
AU2017343752B2 (en) 2016-10-14 2021-12-16 Neomatrix Therapeutics, Inc Peptides derived from fibronectin with improved bioactivity and reduced susceptibility to neutrophil elastase degradation
US10550185B2 (en) 2016-10-14 2020-02-04 Xencor, Inc. Bispecific heterodimeric fusion proteins containing IL-15-IL-15Rα Fc-fusion proteins and PD-1 antibody fragments
TW202300515A (en) 2016-10-20 2023-01-01 法商賽諾菲公司 Anti-chikv antibodies and uses thereof
WO2018081318A1 (en) 2016-10-25 2018-05-03 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Prefusion coronavirus spike proteins and their use
US11414482B2 (en) 2016-11-08 2022-08-16 University Of Miami Anti-secretogranin III (SCG3) antibodies and uses thereof
EP3541847A4 (en) 2016-11-17 2020-07-08 Seattle Genetics, Inc. Glycan-interacting compounds and methods of use
KR102580647B1 (en) 2016-12-07 2023-09-20 몰레큘러 템플레이츠, 인코퍼레이션. Shiga toxin A subunit effector polypeptides, Shiga toxin effector scaffolds, and cell-targeting molecules for site-specific conjugation
CN110087668A (en) 2016-12-07 2019-08-02 Ra制药公司 The regulator of complement activity
AU2018210404B2 (en) 2017-01-19 2022-01-27 Novo Nordisk A/S ApoC-II mimetic peptides
JOP20190189A1 (en) 2017-02-02 2019-08-01 Amgen Res Munich Gmbh Low ph pharmaceutical composition comprising t cell engaging antibody constructs
WO2018152496A1 (en) 2017-02-17 2018-08-23 The Usa, As Represented By The Secretary, Dept. Of Health And Human Services Compositions and methods for the diagnosis and treatment of zika virus infection
MA47812A (en) 2017-03-03 2021-04-14 Seagen Inc COMPOUNDS INTERACTING WITH GLYCAN AND METHODS OF USE
GB201703876D0 (en) 2017-03-10 2017-04-26 Berlin-Chemie Ag Pharmaceutical combinations
CN110382544B (en) 2017-03-16 2023-12-22 先天制药公司 Compositions and methods for treating cancer
WO2018183173A1 (en) 2017-03-27 2018-10-04 Boehringer Ingelheim International Gmbh Anti il-36r antibodies combination therapy
US10729741B2 (en) 2017-03-27 2020-08-04 Neomatrix Therapeutics Inc. Methods of treating burns with i.v. cP12 in a window from 2 to 6 hours after injury
WO2018200742A1 (en) 2017-04-25 2018-11-01 The Usa, As Represented By The Secretary, Dept. Of Health And Human Services Antibodies and methods for the diagnosis and treatment of epstein barr virus infection
UY37726A (en) 2017-05-05 2018-11-30 Amgen Inc PHARMACEUTICAL COMPOSITION THAT INCLUDES BISPECTIFIC ANTIBODY CONSTRUCTIONS FOR IMPROVED STORAGE AND ADMINISTRATION
US10793634B2 (en) 2017-06-09 2020-10-06 Boehringer Ingelheim International Gmbh Anti-TrkB antibodies
WO2019006472A1 (en) 2017-06-30 2019-01-03 Xencor, Inc. Targeted heterodimeric fc fusion proteins containing il-15/il-15ra and antigen binding domains
EP3648788A1 (en) 2017-07-07 2020-05-13 Baxalta Incorporated Treatment of gastrointestinal bleeding in patients with severe von willebrand disease by administration of recombinant vwf
BR112020000322A2 (en) 2017-07-07 2020-07-14 Baxalta Incorporated use of recombinant von willebrand factor (rvwf)
EP3655430A1 (en) 2017-07-19 2020-05-27 The U.S.A. as represented by the Secretary, Department of Health and Human Services Antibodies and methods for the diagnosis and treatment of hepatitis b virus infection
CA3073537A1 (en) 2017-08-22 2019-02-28 Sanabio, Llc Soluble interferon receptors and uses thereof
EP3704154A1 (en) 2017-11-02 2020-09-09 Oxford BioTherapeutics Ltd Antibodies and methods of use
US10981992B2 (en) 2017-11-08 2021-04-20 Xencor, Inc. Bispecific immunomodulatory antibodies that bind costimulatory and checkpoint receptors
CA3082383A1 (en) 2017-11-08 2019-05-16 Xencor, Inc. Bispecific and monospecific antibodies using novel anti-pd-1 sequences
MX2020004933A (en) 2017-11-14 2021-01-08 Arcellx Inc D-domain containing polypeptides and uses thereof.
EP3724229A1 (en) 2017-12-11 2020-10-21 Amgen Inc. Continuous manufacturing process for bispecific antibody products
CN111655718A (en) 2017-12-19 2020-09-11 Xencor股份有限公司 Engineered IL-2 FC fusion proteins
UY38041A (en) 2017-12-29 2019-06-28 Amgen Inc CONSTRUCTION OF BIESPECFIC ANTIBODY DIRECTED TO MUC17 AND CD3
EP3732193A1 (en) 2017-12-29 2020-11-04 Alector LLC Anti-tmem106b antibodies and methods of use thereof
WO2019136158A1 (en) 2018-01-03 2019-07-11 Spitfire Pharma, Inc. Improved peptide pharmaceuticals for treatment of nash and other disorders
KR20200115546A (en) 2018-01-26 2020-10-07 제넨테크, 인크. IL-22 Fc fusion protein and method of use
BR112020015016A2 (en) 2018-01-26 2020-12-29 Genentech, Inc. PHARMACEUTICAL COMPOSITIONS, METHODS OF TREATMENT OF INFLAMMATORY INTESTINAL DISEASE, INHIBITION OF MICROBIAL INFECTION IN THE INTESTINE AND ACCELERATING OR IMPROVING WOUND HEALING
EP3746476A1 (en) 2018-01-31 2020-12-09 Alector LLC Anti-ms4a4a antibodies and methods of use thereof
BR112020016400A2 (en) 2018-02-14 2020-12-15 Viela Bio, Inc. ANTIBODIES FOR THYROSINE KINASE 3 RECEPTOR BINDER SIMILAR TO MCDONOUGH FELINE SARCOMA (FMS) (FLT3L) AND THEIR USES FOR THE TREATMENT OF AUTOIMMUNE AND INFLAMMATORY DISEASES
AU2019226085A1 (en) 2018-02-21 2020-09-17 Genentech, Inc. Dosing for treatment with IL-22 Fc fusion proteins
US11485782B2 (en) 2018-03-14 2022-11-01 Beijing Xuanyi Pharmasciences Co., Ltd. Anti-claudin 18.2 antibodies
CN117126279A (en) 2018-03-20 2023-11-28 鸿运华宁(杭州)生物医药有限公司 GIPR antibody and fusion protein of GIPR antibody and GLP-1, and pharmaceutical composition and application thereof
SG11202009213TA (en) 2018-03-21 2020-10-29 Baxalta Inc Separation of vwf and vwf propeptide by chromatographic methods
US10982006B2 (en) 2018-04-04 2021-04-20 Xencor, Inc. Heterodimeric antibodies that bind fibroblast activation protein
WO2019195561A2 (en) 2018-04-06 2019-10-10 BioLegend, Inc. Anti-tetraspanin 33 agents and compositions and methods for making and using the same
CN110357959B (en) 2018-04-10 2023-02-28 鸿运华宁(杭州)生物医药有限公司 GCGR antibody, fusion protein of GCGR antibody and GLP-1, and pharmaceutical composition and application of GCGR antibody and fusion protein
WO2019204655A1 (en) 2018-04-18 2019-10-24 Xencor, Inc. Tim-3 targeted heterodimeric fusion proteins containing il-15/il-15ra fc-fusion proteins and tim-3 antigen binding domains
JP2021521784A (en) 2018-04-18 2021-08-30 ゼンコア インコーポレイテッド PD-1 targeted heterodimer fusion proteins containing IL-15 / IL-15RaFc fusion proteins and PD-1 antigen binding domains and their use
JP2021522286A (en) 2018-04-30 2021-08-30 メディミューン リミテッド Conjugate to target and clear aggregates
PE20210419A1 (en) 2018-04-30 2021-03-08 Takeda Pharmaceuticals Co CANNABINOID TYPE 1 (CB1) RECEPTOR BINDING PROTEINS AND USES OF THEM
EP3788071A1 (en) 2018-05-02 2021-03-10 The United States Of America, As Represented By The Secretary, Department of Health and Human Services Antibodies and methods for the diagnosis, prevention, and treatment of epstein barr virus infection
UA128113C2 (en) 2018-05-25 2024-04-10 ЕЛЕКТОР ЕлЕлСі Anti-sirpa antibodies and methods of use thereof
CN110655577A (en) 2018-06-13 2020-01-07 鸿运华宁(杭州)生物医药有限公司 APJ antibody and fusion protein thereof with Elabela, and pharmaceutical composition and application thereof
GB201809746D0 (en) 2018-06-14 2018-08-01 Berlin Chemie Ag Pharmaceutical combinations
BR112020024412A8 (en) 2018-06-18 2023-03-21 Innate Pharma ANTIBODIES, PHARMACEUTICAL COMPOSITION, KIT, NUCLEIC ACID, HOST CELL, METHODS OF TREATMENT OR PREVENTION OF CANCER, OF REDUCING ACTIVITY, OF INCREASE OF ACTIVITY AND OF INCREASE OF ACTIVATION
AU2019291890A1 (en) 2018-06-29 2020-12-17 Boehringer Ingelheim International Gmbh Anti-CD40 antibodies for use in treating autoimmune disease
CA3099176A1 (en) 2018-06-29 2020-01-02 Alector Llc Anti-sirp-beta1 antibodies and methods of use thereof
JOP20200343A1 (en) 2018-07-02 2020-12-31 Xencor Inc Anti-steap1 antigen-binding protein
LT3618928T (en) 2018-07-13 2023-04-11 Alector Llc Anti-sortilin antibodies and methods of use thereof
WO2020021061A1 (en) 2018-07-26 2020-01-30 Pieris Pharmaceuticals Gmbh Humanized anti-pd-1 antibodies and uses thereof
WO2020025532A1 (en) 2018-07-30 2020-02-06 Amgen Research (Munich) Gmbh Prolonged administration of a bispecific antibody construct binding to cd33 and cd3
CA3107192A1 (en) 2018-08-03 2020-02-06 Amgen Research (Munich) Gmbh Antibody constructs for cldn18.2 and cd3
JP2021534769A (en) 2018-08-31 2021-12-16 エーエルエックス オンコロジー インコーポレイテッド Decoy polypeptide
CN113164597A (en) 2018-09-24 2021-07-23 爱尔皮奥制药公司 Multispecific antibodies targeting HPTP-beta (VE-PTP) and VEGF
CN113365697A (en) 2018-09-25 2021-09-07 百进生物科技公司 anti-TLR9 agents and compositions and methods of making and using the same
CA3114295A1 (en) 2018-09-28 2020-04-02 Kyowa Kirin Co., Ltd. Il-36 antibodies and uses thereof
SG11202103192RA (en) 2018-10-03 2021-04-29 Xencor Inc Il-12 heterodimeric fc-fusion proteins
CA3114802A1 (en) 2018-10-11 2020-04-16 Amgen Inc. Downstream processing of bispecific antibody constructs
KR20210113261A (en) 2019-01-04 2021-09-15 리졸브 테라퓨틱스, 엘엘씨 Treatment of Sjogren's Disease Using Nuclease Fusion Proteins
CN116063520A (en) 2019-01-30 2023-05-05 真和制药有限公司 anti-GAL 3 antibodies and uses thereof
MX2021009114A (en) 2019-02-01 2021-10-13 Takeda Pharmaceuticals Co Methods of prophylactic treatment using recombinant vwf (rvwf).
EP3693023A1 (en) 2019-02-11 2020-08-12 Sanofi Use of anti-ceacam5 immunoconjugates for treating lung cancer
MX2021009514A (en) 2019-02-07 2021-11-04 Sanofi Sa Use of anti-ceacam5 immunoconjugates for treating lung cancer.
KR20210134725A (en) 2019-03-01 2021-11-10 젠코어 인코포레이티드 Heterodimeric Antibodies that Bind to ENPP3 and CD3
AU2020248645A1 (en) 2019-03-27 2021-10-28 Tigatx, Inc. Engineered IgA antibodies and methods of use
CN114040800A (en) 2019-04-09 2022-02-11 阿波科有限责任公司 Killer cell lectin-like receptor subfamily G member 1(KLRG1) depleting antibodies
CA3130449A1 (en) 2019-04-30 2020-11-05 Gigagen, Inc. Recombinant polyclonal proteins and methods of use thereof
CA3137377A1 (en) 2019-05-09 2020-11-12 Boehringer Ingelheim International Gmbh Anti-sema3a antibodies and their uses for treating eye or ocular diseases
TW202045711A (en) 2019-06-13 2020-12-16 美商安進公司 Automated biomass-based perfusion control in the manufacturing of biologics
TW202115112A (en) 2019-06-27 2021-04-16 德商百靈佳殷格翰國際股份有限公司 Anti-angpt2 antibodies
BR112021024997A2 (en) 2019-07-03 2022-01-25 Oxford Biotherapeutics Ltd Antibodies, polynucleotide, expression vector, host cell, pharmaceutical composition, methods and use
CN112239507A (en) 2019-07-17 2021-01-19 鸿运华宁(杭州)生物医药有限公司 Fusion protein of ETA antibody and TGF-beta Trap, and pharmaceutical composition and application thereof
CN112300279A (en) 2019-07-26 2021-02-02 上海复宏汉霖生物技术股份有限公司 Methods and compositions directed to anti-CD 73 antibodies and variants
KR20220058540A (en) 2019-07-31 2022-05-09 알렉터 엘엘씨 Anti-MS4A4A antibodies and methods of use thereof
US20210032370A1 (en) 2019-08-02 2021-02-04 Immatics Biotechnologies Gmbh Recruiting agent further binding an mhc molecule
DE102019121007A1 (en) 2019-08-02 2021-02-04 Immatics Biotechnologies Gmbh Antigen binding proteins that specifically bind to MAGE-A
US20220281967A1 (en) 2019-08-02 2022-09-08 Orega Biotech Novel il-17b antibodies
US20220289807A1 (en) 2019-08-13 2022-09-15 Amgen Inc. Interleukin-2 muteins for the expansion of t-regulatory cells
MX2022002981A (en) 2019-09-10 2022-04-06 Amgen Inc Purification method for bispecific antigen-binding polypeptides with enhanced protein l capture dynamic binding capacity.
JP2022547556A (en) 2019-09-11 2022-11-14 武田薬品工業株式会社 Therapies involving the complex of von Willebrand factor and complement C1Q
CN112521501A (en) 2019-09-18 2021-03-19 鸿运华宁(杭州)生物医药有限公司 GIPR antibody and fusion protein thereof with GLP-1, and pharmaceutical composition and application thereof
TW202126685A (en) 2019-09-24 2021-07-16 德商百靈佳殷格翰國際股份有限公司 Anti-nrp1a antibodies and their uses for treating eye or ocular diseases
CN114667298A (en) 2019-11-04 2022-06-24 艾利妥 SIGLEC-9ECD fusion molecules and methods of use thereof
AU2020381536A1 (en) 2019-11-13 2022-04-21 Amgen Inc. Method for reduced aggregate formation in downstream processing of bispecific antigen-binding molecules
AU2020397416A1 (en) 2019-12-03 2022-07-14 Evotec International Gmbh Interferon-associated antigen binding proteins for use in treating hepatitis B infection
CN115052625A (en) 2019-12-03 2022-09-13 埃沃特克国际有限责任公司 Interferon-associated antigen binding proteins and uses thereof
JP2023505256A (en) 2019-12-05 2023-02-08 ソレント・セラピューティクス・インコーポレイテッド Compositions and methods comprising anti-CD47 antibodies in combination with tumor-targeting antibodies
CR20220329A (en) 2019-12-13 2022-11-23 Alector Llc Anti-mertk antibodies and methods of use thereof
IL293471A (en) 2019-12-17 2022-08-01 Amgen Inc Dual interleukin-2 /tnf receptor agonist for use in therapy
EP4090365A1 (en) 2020-01-15 2022-11-23 Immatics Biotechnologies GmbH Antigen binding proteins specifically binding prame
CN115427447A (en) 2020-01-17 2022-12-02 百进生物科技公司 anti-TLR 7 agents and compositions and methods of making and using the same
US20230093169A1 (en) 2020-01-22 2023-03-23 Amgen Research (Munch) Gmbh Combinations of antibody constructs and inhibitors of cytokine release syndrome and uses thereof
AU2021216945A1 (en) 2020-02-04 2022-09-01 Takeda Pharmaceutical Company Limited Treatment of menorrhagia in patients with severe von Willebrand Disease by administration of recombinant VWF
US20230183377A1 (en) 2020-02-26 2023-06-15 Sorrento Therapeutics, Inc. Activatable antigen binding proteins with universal masking moieties
EP4118113A1 (en) 2020-03-12 2023-01-18 Amgen Inc. Method for treatment and prophylaxis of crs in patients comprising a combination of bispecifc antibodies binding to cds x cancer cell and tnfalpha or il-6 inhibitor
WO2021195089A1 (en) 2020-03-23 2021-09-30 Sorrento Therapeutics, Inc. Fc-coronavirus antigen fusion proteins, and nucleic acids, vectors, compositions and methods of use thereof
CN116075525A (en) 2020-03-31 2023-05-05 艾莱克特有限责任公司 anti-MERTK antibodies and methods of use thereof
WO2021207662A1 (en) 2020-04-10 2021-10-14 Genentech, Inc. Use of il-22fc for the treatment or prevention of pneumonia, acute respiratory distress syndrome, or cytokine release syndrome
CA3180222A1 (en) 2020-04-15 2021-10-21 Voyager Therapeutics, Inc. Tau binding compounds
EP4138884A1 (en) 2020-04-20 2023-03-01 Sorrento Therapeutics, Inc. Pulmonary administration of ace2 polypeptides
WO2021231976A1 (en) 2020-05-14 2021-11-18 Xencor, Inc. Heterodimeric antibodies that bind prostate specific membrane antigen (psma) and cd3
AU2021275049A1 (en) 2020-05-19 2022-12-22 Amgen Inc. MAGEB2 binding constructs
CA3181776A1 (en) 2020-05-26 2021-12-02 Boehringer Ingelheim International Gmbh Anti-pd-1 antibodies
JP2023527972A (en) 2020-05-29 2023-07-03 アムジエン・インコーポレーテツド Reduced Adverse Effect Administration of Bispecific Constructs that Bind CD33 and CD3
CA3177152A1 (en) 2020-06-12 2021-12-16 David Scott Johnson Recombinant polyclonal proteins targeting covid-19 and methods of use thereof
WO2021259227A1 (en) 2020-06-23 2021-12-30 江苏康缘药业股份有限公司 Anti-cd38 antibody and use thereof
EP4172207A1 (en) 2020-06-26 2023-05-03 Sorrento Therapeutics, Inc. Anti-pd1 antibodies and uses thereof
CA3165342A1 (en) 2020-06-29 2022-01-06 James Arthur Posada Treatment of sjogren's syndrome with nuclease fusion proteins
WO2022031834A1 (en) 2020-08-05 2022-02-10 Gigagen, Inc. Recombinant polyclonal proteins targeting zika and methods of use thereof
KR20230062600A (en) 2020-09-04 2023-05-09 메르크 파텐트 게엠베하 Anti-CEACAM5 Antibodies and Conjugates and Uses Thereof
US20220089759A1 (en) 2020-09-21 2022-03-24 Boehringer Ingelheim International Gmbh Use of anti-cd40 antibodies for treatment of inflammatory conditions
WO2022081870A1 (en) 2020-10-14 2022-04-21 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Stabilized norovirus virus-like particles as vaccine immunogens
US20220127344A1 (en) 2020-10-23 2022-04-28 Boehringer Ingelheim International Gmbh Anti-sema3a antibodies and their uses for treating a thrombotic disease of the retina
WO2022093641A1 (en) 2020-10-30 2022-05-05 BioLegend, Inc. Anti-nkg2a agents and compositions and methods for making and using the same
WO2022093640A1 (en) 2020-10-30 2022-05-05 BioLegend, Inc. Anti-nkg2c agents and compositions and methods for making and using the same
CN116635421A (en) 2020-11-06 2023-08-22 安进公司 Polypeptide constructs that bind CD3
KR20230098335A (en) 2020-11-06 2023-07-03 암젠 인크 Antigen binding domains with reduced clipping ratio
UY39508A (en) 2020-11-06 2022-05-31 Amgen Res Munich Gmbh BSPECIFIC ANTIGEN-BINDING MOLECULES WITH MULTIPLE TARGETS OF ENHANCED SELECTIVITY
BR112023008629A2 (en) 2020-11-06 2023-10-03 Amgen Inc POLYPEPTIDE CONSTRUCTS THAT SELECTIVELY BIND CLDN6 AND CD3
WO2022132923A1 (en) 2020-12-16 2022-06-23 Voyager Therapeutics, Inc. Tau binding compounds
WO2022147463A2 (en) 2020-12-31 2022-07-07 Alamar Biosciences, Inc. Binder molecules with high affinity and/ or specificity and methods of making and use thereof
JP2024504696A (en) 2021-01-20 2024-02-01 バイオアントレ エルエルシー CTLA4-binding proteins and methods of treating cancer
EP4288457A2 (en) 2021-02-05 2023-12-13 Boehringer Ingelheim International GmbH Anti-il1rap antibodies
IL305736A (en) 2021-03-09 2023-11-01 Xencor Inc Heterodimeric antibodies that bind cd3 and cldn6
EP4305065A1 (en) 2021-03-10 2024-01-17 Xencor, Inc. Heterodimeric antibodies that bind cd3 and gpc3
JP2024510435A (en) 2021-03-18 2024-03-07 シージェン インコーポレイテッド Selective drug release from internalization complexes of bioactive compounds
JP2024512002A (en) 2021-03-18 2024-03-18 アレクトル エルエルシー Anti-TMEM106B antibody and method of use thereof
WO2022204274A1 (en) 2021-03-23 2022-09-29 Alector Llc Anti-tmem106b antibodies for treating and preventing coronavirus infections
WO2022204529A1 (en) 2021-03-26 2022-09-29 Abcuro, Inc. Anti-klrg1 antibodies
BR112023019512A2 (en) 2021-03-26 2023-10-31 Abcuro Inc ANTI-KLRG1 ANTIBODIES
CN115141276A (en) 2021-03-31 2022-10-04 鸿运华宁(杭州)生物医药有限公司 Antibody capable of being specifically combined with human endothelin receptor and application thereof in treatment of diabetic nephropathy and chronic nephropathy
WO2022212836A1 (en) 2021-04-01 2022-10-06 Pyxis Oncology, Inc. Gpnmb antibodies and methods of use
CA3215594A1 (en) 2021-04-02 2022-10-06 Agnieszka KIELCZEWSKA Mageb2 binding constructs
CN117279947A (en) 2021-05-06 2023-12-22 安进研发(慕尼黑)股份有限公司 Antigen binding molecules targeting CD20 and CD22 for use in proliferative diseases
JP7436711B2 (en) 2021-06-04 2024-02-22 ベーリンガー インゲルハイム インターナショナル ゲゼルシャフト ミット ベシュレンクテル ハフツング Anti-SIRP-alpha antibody
WO2022261183A2 (en) 2021-06-08 2022-12-15 Dana-Farber Cancer Institute, Inc. Compositions and methods for treating and/or identifying an agent for treating intestinal cancers
EP4351732A1 (en) 2021-06-09 2024-04-17 Evotec International GmbH Interferon-associated antigen binding proteins for use for the treatment or prevention of coronavirus infection
EP4355783A1 (en) 2021-06-16 2024-04-24 Alector LLC Monovalent anti-mertk antibodies and methods of use thereof
CN117642426A (en) 2021-06-16 2024-03-01 艾莱克特有限责任公司 Bispecific anti-MerTK and anti-PDL 1 antibodies and methods of use thereof
AU2022320627A1 (en) 2021-07-26 2024-02-08 Abcuro, Inc. Killer cell lectin-like receptor subfamily g member 1 (klrg1) depleting antibodies
WO2023069919A1 (en) 2021-10-19 2023-04-27 Alector Llc Anti-cd300lb antibodies and methods of use thereof
WO2023097119A2 (en) 2021-11-29 2023-06-01 Dana-Farber Cancer Institute, Inc. Methods and compositions to modulate riok2
WO2023099682A1 (en) 2021-12-02 2023-06-08 Sanofi Ceacam5 adc–anti-pd1/pd-l1 combination therapy
TW202339804A (en) 2021-12-02 2023-10-16 法商賽諾菲公司 Cea assay for patient selection in cancer therapy
AR128065A1 (en) 2021-12-22 2024-03-20 Cdr Life Ag ANTI-C3 ANTIBODIES AND ANTIGEN-BINDING FRAGMENTS THEREOF AND THEIR USES TO TREAT OPHTHALMIC OR EYE DISEASES
WO2023131901A1 (en) 2022-01-07 2023-07-13 Johnson & Johnson Enterprise Innovation Inc. Materials and methods of il-1beta binding proteins
WO2023172968A1 (en) 2022-03-09 2023-09-14 Merck Patent Gmbh Anti-gd2 antibodies, immunoconjugates and therapeutic uses thereof
WO2023170239A1 (en) 2022-03-09 2023-09-14 Merck Patent Gmbh Methods and tools for conjugation to antibodies
TW202346368A (en) 2022-05-12 2023-12-01 德商安美基研究(慕尼黑)公司 Multichain multitargeting bispecific antigen-binding molecules of increased selectivity
WO2023240287A1 (en) 2022-06-10 2023-12-14 Bioentre Llc Combinations of ctla4 binding proteins and methods of treating cancer
WO2023250388A1 (en) 2022-06-22 2023-12-28 Voyager Therapeutics, Inc. Tau binding compounds
WO2024013727A1 (en) 2022-07-15 2024-01-18 Janssen Biotech, Inc. Material and methods for improved bioengineered pairing of antigen-binding variable regions
WO2024020051A1 (en) 2022-07-19 2024-01-25 BioLegend, Inc. Anti-cd157 antibodies, antigen-binding fragments thereof and compositions and methods for making and using the same
WO2024026447A1 (en) 2022-07-29 2024-02-01 Alector Llc Anti-gpnmb antibodies and methods of use thereof
WO2024040114A2 (en) 2022-08-18 2024-02-22 BioLegend, Inc. Anti-axl antibodies, antigen-binding fragments thereof and methods for making and using the same
WO2024059675A2 (en) 2022-09-14 2024-03-21 Amgen Inc. Bispecific molecule stabilizing composition

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4179337A (en) * 1973-07-20 1979-12-18 Davis Frank F Non-immunogenic polypeptides
SU1022988A1 (en) * 1979-09-28 1983-06-15 Всесоюзный кардиологический научный центр АМН СССР Stabilized urokinase having trombolite activity and method of producing same

Also Published As

Publication number Publication date
US4640835A (en) 1987-02-03

Similar Documents

Publication Publication Date Title
CA1217718A (en) Plasminogen activator derivatives
US4495285A (en) Plasminogen activator derivatives
EP0041766B1 (en) New plasminogen activator and pharmaceutical composition having thrombolytic activity
US5407673A (en) Fibrinolysis and fibrinogenolysis treatment
EP0123304B1 (en) A method for stabilizing tissue plasminogen activator and a stable aqueous solution or powder containing the same
EP0100982A2 (en) Novel purified plasminogen activator, process for its production and thrombolytic composition containing it
EP0620738A1 (en) Fibrinolysis and fibrinogenolysis treatment with plasmin
Smith et al. Acyl-enzymes as thrombolytic agents in a rabbit model of venous thrombosis
EP0217379A2 (en) Thrombolytic composition and a process for production thereof
KR970005837B1 (en) Process for preparing a solution with a high specific volume activity of a protein with a tissue plasminogen activator activity(t-pa), solution containing the protein with t-pa activity and fibinolytic composition containing this solution
JPS61243024A (en) Novel compound, manufacture and medicinal composition
US5491129A (en) Synthetic peptides derived from vitronectin and pharmaceutical compositions comprising them
US4082612A (en) Plasminogen activator complex
US4996050A (en) Fibrinolytic activity enhancer
US4507283A (en) Pharmacologically active compounds
EP0151593A1 (en) Enzyme derivatives.
KR0137476B1 (en) Factor ñ˜ñ for the prevention intraventricular hemorrhage
CA1283047C (en) Stabilized plasminogen activator precursor and method of producing the same
BANG Physiology and biochemistry of fibrinolysis
JPS6360938A (en) Plasminogen activator of modified tissue type and production thereof
US20060147441A1 (en) Plasminogen fragment having activity to inhibit tumor metastasis and growth and process for preparing same technical field
JP3806471B2 (en) Plasminogen fragment having tumor metastasis growth inhibitory effect and method for preparing the fragment
JPS59167520A (en) Novel plasminogen activator derivative, its preparation and pharmaceutical containing the same
Breddin Perspectives of the Clinical use of Hirudin
JPH0769921A (en) Agent for preventing and treating vasa sanguinea retinae occlusion

Legal Events

Date Code Title Description
MKEX Expiry