WO1994000156A1 - Polymer-bound paclitaxel derivatives - Google Patents

Polymer-bound paclitaxel derivatives Download PDF

Info

Publication number
WO1994000156A1
WO1994000156A1 PCT/EP1993/001433 EP9301433W WO9400156A1 WO 1994000156 A1 WO1994000156 A1 WO 1994000156A1 EP 9301433 W EP9301433 W EP 9301433W WO 9400156 A1 WO9400156 A1 WO 9400156A1
Authority
WO
WIPO (PCT)
Prior art keywords
phe
gly
leu
βala
ala
Prior art date
Application number
PCT/EP1993/001433
Other languages
French (fr)
Inventor
Nicola Mongelli
Francesco Angelucci
Enrico Pesenti
Antonio Suarato
Giovanni Biasoli
Original Assignee
Pharmacia Carlo Erba S.R.L.
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 to EP93912905A priority Critical patent/EP0600062B1/en
Application filed by Pharmacia Carlo Erba S.R.L. filed Critical Pharmacia Carlo Erba S.R.L.
Priority to KR1019940700488A priority patent/KR100281606B1/en
Priority to PL93302437A priority patent/PL173898B1/en
Priority to DE69330776T priority patent/DE69330776T2/en
Priority to RU94016158A priority patent/RU2130462C1/en
Priority to AT93912905T priority patent/ATE205725T1/en
Priority to AU43233/93A priority patent/AU659750B2/en
Priority to CZ94620A priority patent/CZ62094A3/en
Priority to JP50198194A priority patent/JP3693340B2/en
Priority to UA94005444A priority patent/UA39926C2/en
Publication of WO1994000156A1 publication Critical patent/WO1994000156A1/en
Priority to FI940733A priority patent/FI940733A0/en
Priority to NO940567A priority patent/NO940567L/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D305/00Heterocyclic compounds containing four-membered rings having one oxygen atom as the only ring hetero atoms
    • C07D305/14Heterocyclic compounds containing four-membered rings having one oxygen atom as the only ring hetero atoms condensed with carbocyclic rings or ring systems
    • 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
    • A61K47/58Medicinal 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 by reactions only involving carbon-to-carbon unsaturated bonds, e.g. poly[meth]acrylate, polyacrylamide, polystyrene, polyvinylpyrrolidone, polyvinylalcohol or polystyrene sulfonic acid resin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents

Definitions

  • POLYMER-BOUND PACLITAXEL DERIVATIVES The present invention is directed to polymer-bound paclitaxel and polymer-bound paclitaxel derivatives endowed with antitumor activity, to a method for their preparation and to pharmaceutical compositions containing them.
  • Paclitaxel (also named Taxol in several publications) is a member of the taxane family of diterpenes, isolated and characterized from an extract of bark of Taxus brevifolia L. ; other analogues of paclitaxel are also known and were prepared by semisynthesis starting from 10-deacetyl baccatin III, extracted from the needles of Taxus baccata L.. see Wain et al. in JACS, .93 . , 2325 (1971) and Lovelle et al., Proc. Am. Assoc. Cancer Res., 31. p. 417, (1990) .
  • the present invention provides a polymer conjugate of the formula I consisting essentially of: from 90 to 99.9 mol % of units of the formula:
  • R is a phenyl or t-butoxy group
  • R 3 is H or an acetyl group
  • a and A which may be the same or different, represent a chemical single bond, an amino acid residue or peptide spacer selected from ⁇ Ala, Gly, Phe-Gly, Phe-Phe-, Leu-Gly, Val-Ala, Phe-Ala, Leu- Phe, Leu-Ala, Phe-Leu-Gly, Phe-Phe-Leu, Leu-Leu-Gly, Phe- Tyr-Ala, Phe-Gly-Phe, Ph -Phe-Gly, Phe-Leu-Gly-Phe, Gly- Phe-Leu-Gly-Phe,Gly-3Ala, Phe-Gly- ⁇ Ala, Phe-Phe- ⁇ Ala, Leu-Gly- ⁇ Ala, Val-Ala- ⁇ Ala, Phe-Ala- ⁇ Ala, Leu-Phe- ⁇ A
  • the present invention provides polymer conjugates of paclitaxel and derivatives of paclitaxel with improved water solubility and decreased toxicity.
  • the mol % of units containing the paclitaxel and paclitaxel derivatives is from 0.5 to 2 , more preferably, the content of paclitaxel in the polymer was from 2 to 10 % (w/w) , most preferred compounds are those characterized by a content of from 4 to 7 % (w/w) .
  • the wavy line denotes that the oxygen linked at position 7 of the paclitaxel structure may be in both configurations, i.e. ⁇ (natural) or ⁇ .
  • R represents a phenyl group
  • R 3 is an acetyl group
  • A is a Phe-Leu-Gly or Phe-Leu-Gly- ⁇ Ala residue. All the amino acid residues have the natural L- configuration.
  • the polymer conjugate is a copolymer of 1-methacryloylamino- 2-hydroxypropane, (methacryloylglycyl-phenylalanyl- leucylglycyl) 3-amino-2 hydroxypropane and 2'- (methacryloylglycylphenylalanylleucylglycyl-/3alanyl) paclitaxel.
  • Samples were extracted by adding 75 ⁇ l of Tetra Butyl Ammonium Phosphate (TBAP) 0.5 M, 1250 ⁇ l CH 3 CN and 150 ⁇ l of NaCl 5M and vigorously shacked for 20' at 4°C. After that time samples were spun at 15000 x g for 10' and the supernatants were collected and evaporated using a high vacuum centrifuge . Samples were recovered by adding 500 ⁇ l of MeOH:H 2 0 (75:25 V/V) and injected into HPLC for determining the total paclitaxel percentage content.
  • TBAP Tetra Butyl Ammonium Phosphate
  • the present invention also provides a process for preparing a polymer conjugate which process comprises reacting a compound of the formula II
  • a 2 and A 3 are a chemical bond and the other one is A, and A, R and R 3 are as defined above, with an activated polymer consisting essentially of from 90 to 99.9 mol % of units represented by formula
  • reaction between the compounds of the formula II and the activated polymer is carried out in an anhydrous polar organic solvent such as dimethylsulfoxide or dimethylformamide optionally in presence of an organic or inorganic base such as an alkaline carbonate, dimethyla inopyridine or triethylamine.
  • an organic or inorganic base such as an alkaline carbonate, dimethyla inopyridine or triethylamine.
  • the reaction is typically carried out at a temperature of from 15 to 40°C, preferably at room temperature.
  • the alkaline carbonate is, for example, an alkali metal carbonate or an alkaline earth metal carbonate.
  • 7-epi derivatives may be prepared by refluxing in toluene paclitaxel or its analogs in the presence of a base (Na 2 C0 3 or diazabicycloundecene)
  • a base Na 2 C0 3 or diazabicycloundecene
  • Other compounds of the formula II are new, in particular those in which A 3 is ⁇ hla residue and those in which either A 2 or A 3 represents di, tri, or tetra peptide spacer as defined above for A and are within the scope of the invention.
  • the compounds of formula II wherein A 2 is not a chemical single bond may be prepared by reacting a paclitaxel or a paclitaxel analog with protected amino acid or peptide in the presence of a condensating reagent, and with or without the additional presence of a catalyst, preferably at room temperature, followed by the removal of the protecting group with known methods.
  • the condensation may be also carried out using activated esters such as paranitrophenyl ester of peptide or amino acid.
  • Suitable condensing reagents include carbodiimides such as dicyclohexyl carbodii ide (DCC) .
  • Suitable catalysts include 4-dimethylamino-pyridine (DMAP) , pyridine or triethylamine.
  • Various known amino protecting groups can be utilized and commercially available protected amino acid or peptide ⁇ can be utilized as the starting materials.
  • Amino acids or peptides protected with t-BOC, trityl, FMOC or carbobenzyloxy (CBZ) can be utilized.
  • Amino Acid or peptides procted with t-BOC, trityl or FMOC groups are preferred.
  • the compounds of the formula II wherein A 3 is not a chemical single bond may be prepared by protecting or blocking the 2'-hydroxy group and then esterifying the 7- position hydroxyl and then removing the 2'-protecting or blocking group.
  • the compounds of the formula II wherein A 3 is Gly or Ala residue are prepared by reacting paclitaxel with 2-3 equivalents of N-protected amino acid to produce 2' ,7-disubstituted paclitaxel, the 2'-position amino acid is cleaved and then the 7-position amino acid is deprotected.
  • Reaction of paclitaxel and the protected amino acid is conducted in the presence of a condensing reagent and a catalyst, like those above defined.
  • Cleavage of the 2'-amino acid is conducted by adjusting the pH of the 2'-7-(amino acid) paclitaxel solution to pH 7-7.4 for example by mixture of the 2' ,7-di(amino acid) paclitaxel in a phosphate buffer at pH 7-7.4 or with a slight excess of NaHC0 3 .
  • Deprotection of the amino acid is conducted under a known amino acid deprotection method, such as mild acid treatment with, for example, acetic acid, or by reduction.
  • paclitaxel is allowed to react with 2- 3 mol. equivalent of N-protected amino acid (t.boc, CBZ or FMOC protected) in methylene chloride in the presence of DCC and a catalytic amount of 4-dimethylaminopyridine.
  • the protected amino acid is introduced at 2' and 7-position.
  • the 2',7-bis amino acid derivative of paclitaxel is allowed to stand in presence of NaHC0 3 in H 2 0/MeOH for 2-5 hours, whereby selective deprotection at the 2'-position occurs to yield the 7-substituted derivative of paclitaxel.
  • the protecting groups are removed by appropriate deprotecting agent (e.g., acid, mild base or hydrogenolysis) .
  • the activated polymer is a synthetic, water soluble polymer prepared by the copolymerization of N'-(2- hydroxypropyl) methacrylamide with p-nitrophenylesters of N-methacryloyl oligopeptides, as described in US-A- 4062831 and US-A-4097470.
  • the polymer conjugates of the formula I and the new paclitaxel derivatives of the formula II exhibit good water solubility, biocompatibility and release the paclitaxel or paclitaxel derivative in the plasma or after internalization into cells by cleaving of the oligopeptide spacers.
  • Biological activity Microtubule assembly and disassembly assay
  • Calf brain tubulin was prepared by two cycles of assembly-disassembly (Shelanski M.L. Gaskin F. and Cantor C.R., Proc. Natl. Acad.Sci. U.S.A. 70, 765-768, 1973) and stored in liquid nitrogen in MAB (0.1 M MES, 2.5 mM EGTA, 0.5 mM MgS0 4 , 0.1 mM EDTA, 0.1 mM DTT pH 6.4) . All the experiments were carried out on protein stored for less than 4 weeks. Before each experiment, the tubulin was kept 30 min at 4°C.
  • copolymer-paclitaxel prepared in Example 6 was tested in vivo against B16 F10 murine melanoma in comparison with paclitaxel.
  • mice C57B16 female mice were obtained from Charles River Italy.
  • paclitaxel was dissolved in a vehicle consisting of polyoxyethylated castor oil (Cremophor EL) 50% and ethanol 50% and then diluted with glucose 5% solution at the desidered concentrations. The solution was slightly hazy and precipitates formation was observed after short time.
  • Cremophor EL polyoxyethylated castor oil
  • the compound of example 6 was easily dissolved in glucose 5% and the resulting solution was clear for long time (more than 2 hours) . Final concentrations were referred to the paclitaxel content of the compound (4% of total) .
  • the murine melanoma B16F10 was used. A suspension of 10 5 tumor cells in 0.2 ml was injected ⁇ ubcutaneously in the flank of mice. Tumor size was measured with caliper and tumor weight was calculated with the formula:
  • Paclitaxel was given intraperitoneally because of its poor solubility and vehicle toxicity.
  • the compound of example 6 was injected intravenously. Both compounds were administered at day 1, 5, 9 after tumor implantation.
  • TOX number of mice which died for toxicity. TOX determination was made when mice died before the control or when significant body weight loss and/or spleen and or/or liver size reduction were observed. From the above data, it can be seen that the polymer conjugates of the present invention exhibit excellent antitumor activity. These compounds, therefore, are useful antitumor agents due to the lower toxicity and increased water solubility as compared to paclitaxel or its derivative. Examples of tumors that can be treated are for istance, sarcomas, carcinoma, lymphomas, neuroblastoma, melanoma, myeloma, Wilms tumor, leukemias and adenocarcinoma.
  • the improved solubility ad decreased toxicity of the polymer-conjugates of the present invention means that they are suitable for intravenous injection or infusion.
  • the dosage depends upon the age, weight and condition of the patient.
  • the dosage may be from 1 mg/kg body weight to 1 g/kg body weight, preferably from 4 to 800 mg/kg body weight.
  • Typical formulations contain a quantity of polymer-bound paclitaxel/poly erbound paclitaxel derivative equivalent to 0.5, 1.5, 10, 20, 25, or 50 mg of the active paclitaxel/paclitaxel derivative.
  • the polymer conjugates may be formulated as pharmaceutical compositions with a pharmaceutically acceptable carrier or diluent. Any appropriate carrier or diluent may be used.
  • the solutions for intravenous injection or infusion may contain as carrier or diluent, for example, sterile water or preferably they may be in the form of sterile, aqueous or isotonic saline solutions.
  • carrier or diluent for example, sterile water or preferably they may be in the form of sterile, aqueous or isotonic saline solutions.
  • reaction solution was quenched with 0.3 ml glacial acetic acid, concentrated under vacuum to a small volume, and then poured into 200 ml of acetone. Ater 30' mixing, the precipitate was filtered and washed with acetone to yield 1.25 g of the title compound.
  • the paclitaxel content was 4.5% (evaluated by enzymatic hydrolysis and HPLC analysis) .
  • the compound 2' (N-Trit-Phe-Leu-Gly)paclitaxel (250 mg) was dissolved into a mixture of glacial acetic (22 ml) and water (6 ml) and the whole was stirred for 1 hour at room temperature. The solvents were evaporated in vacuum to the dryness, the residue stirred with diethylether-hexane 1:1 for 30 minutes and filtered to obtain 160 mg of the title compound.
  • Copolymer of l-methacryloylamino-2-hydroxy -propane, 1(methacryloyl-glycyl)amino-2-hydroxypropane and 2' (methacryloyl-glycyl)paclitaxel To a solution of 1.6 g of copolymer of 1-methacryloyl - amino-2-hydroxypropane and N-(methylen-carbonyl-4-nitro- phenoxy)methacrylamide in 16 ml of anhydrous dimethyl ⁇ formamide were added 100 mg of paclitaxel and 20 mg of dimethylamino-pyridine.
  • the reaction solution was quenched with 0.3 ml of glacial acetic acid, concentrated under vacuum to a small volume and poured in 250 ml of acetone. The mixture was stirred for 1 hour, the precipitate was filtered and washed with acetone to yield 1520 mg of the title compound.
  • the paclitaxel content was 7.8% w/w%.

Abstract

A polymer conjugate consisting essentially of: from 90 to 99.9 mol % of units represented by formula (a); from 0.1 to 5 mol % of units represented by formula (b), wherein one of R1 and R2 is a copolymer residue of formula (c), and the other one is hydrogen atom; from 0 to 9.9 mol % of units represented by formula (d), wherein R is a phenyl or t-butoxy group, R3 is H or an acetyl group, A and A1 which may be the same or different, represent a chemical single bond, an amino acid residue or peptide spacer selected from β Ala, Gly, Phe-Gly, Phe-Phe-, Leu-Gly, Val-Ala, Phe-Ala, Leu-Phe, Leu-Ala, Phe-Leu-Gly, Phe-Phe-Leu, Leu-Leu-Gly, Phe-Tyr-Ala, Phe-Gly-Phe, Phe-Phe-Gly, Phe-Leu-Gly-Phe, Gly-Phe-Leu-Gly-Phe, Gly-βAla, Phe-Gly-βAla, Phe-Phe-βAla, Leu-Gly-βAla, Val-Ala-βAla, Phe-Ala-βAla, Leu-Phe-βAla, Leu-Gly-βAla, Phe-Leu-Gly-βAla, Phe-Phe-Leu βAla, Leu-Leu-Gly-βAla, Phe-Tyr-Ala-βAla, Phe-Gly-Phe-β, Phe-Phe-GlyβAla, Phe-Leu-Gly-Phe-βAla or Gly-Phe-Leu-Gly-Phe-βAla. The compounds are endowed with antitumor activity and show improved water solubility and decreased toxicity in comparison with paclitaxel or its known analogs. A method for their preparation and pharmaceutical compositions containing them are also described.

Description

POLYMER-BOUND PACLITAXEL DERIVATIVES The present invention is directed to polymer-bound paclitaxel and polymer-bound paclitaxel derivatives endowed with antitumor activity, to a method for their preparation and to pharmaceutical compositions containing them.
Paclitaxel (also named Taxol in several publications) is a member of the taxane family of diterpenes, isolated and characterized from an extract of bark of Taxus brevifolia L. ; other analogues of paclitaxel are also known and were prepared by semisynthesis starting from 10-deacetyl baccatin III, extracted from the needles of Taxus baccata L.. see Wain et al. in JACS, .93., 2325 (1971) and Lovelle et al., Proc. Am. Assoc. Cancer Res., 31. p. 417, (1990) . These compounds have been shown to possess potent antitumor activity, but they are very slightly soluble in water and some toxic side-effects are associated also with their administration by injection or intravenous infusion using as carrier cremophor EL (TM) . The present invention provides a polymer conjugate of the formula I consisting essentially of: from 90 to 99.9 mol % of units of the formula:
I CH2
I
CH3— C-CO-NH-CH-CHOH-CH3 from 0.1 to I5 mol % units represented by the formula
Figure imgf000004_0001
wherein one of R, and R2 is a copolymer residue of the formula
CH,
CH3—C-CONH-CH2-CO-A—
and the other one is hydrogen atom; from 0 to 9.9 mol % of units represented by the formula
Figure imgf000004_0002
CH,— C-CONH-CH,-CO-A,-NH-CH,-CHOH-CH,
wherein R is a phenyl or t-butoxy group, R3 is H or an acetyl group, A and A, which may be the same or different, represent a chemical single bond, an amino acid residue or peptide spacer selected from β Ala, Gly, Phe-Gly, Phe-Phe-, Leu-Gly, Val-Ala, Phe-Ala, Leu- Phe, Leu-Ala, Phe-Leu-Gly, Phe-Phe-Leu, Leu-Leu-Gly, Phe- Tyr-Ala, Phe-Gly-Phe, Ph -Phe-Gly, Phe-Leu-Gly-Phe, Gly- Phe-Leu-Gly-Phe,Gly-3Ala, Phe-Gly-βAla, Phe-Phe-βAla, Leu-Gly-βAla, Val-Ala-βAla, Phe-Ala-βAla, Leu-Phe-βAla, Leu-Gly-βAla, Phe-Leu-Gly- Ala, Phe-Phe-Leu βAla, Leu- Leu-Gly-βAla, Phe-Tyr-Ala-βAla, Phe-Gly-Phe-/3, Phe-Phe- Gly-βAla, Phe-Leu-Gly-Phe-βAla or Gly-Phe-Leu-Gly-Phe- jSAla.
More particularly the present invention provides polymer conjugates of paclitaxel and derivatives of paclitaxel with improved water solubility and decreased toxicity. Preferably, the mol % of units containing the paclitaxel and paclitaxel derivatives is from 0.5 to 2 , more preferably, the content of paclitaxel in the polymer was from 2 to 10 % (w/w) , most preferred compounds are those characterized by a content of from 4 to 7 % (w/w) . The wavy line denotes that the oxygen linked at position 7 of the paclitaxel structure may be in both configurations, i.e. β (natural) or α.
Preferably R represents a phenyl group, R3 is an acetyl group and A is a Phe-Leu-Gly or Phe-Leu-Gly-βAla residue. All the amino acid residues have the natural L- configuration. According to a preferred embodiment, the polymer conjugate is a copolymer of 1-methacryloylamino- 2-hydroxypropane, (methacryloylglycyl-phenylalanyl- leucylglycyl) 3-amino-2 hydroxypropane and 2'- (methacryloylglycylphenylalanylleucylglycyl-/3alanyl) paclitaxel.
The % w/w content was determined after enzymatic hydrolysis by HPLC method analogous to that described in Cancer Treatment Rep. 7_1 (1), 1987, p. 53-59. Enzymatic hydrolysis
To 1 ml of urine or human plasmas various concentrations of the polymer-bound paclitaxel were added and at appropriate time (24, 48, 72, 96 h) 100 μl were collected and stored at -70°C until further processing. Extraction procedure
Samples were extracted by adding 75 μl of Tetra Butyl Ammonium Phosphate (TBAP) 0.5 M, 1250 μl CH3CN and 150 μl of NaCl 5M and vigorously shacked for 20' at 4°C. After that time samples were spun at 15000 x g for 10' and the supernatants were collected and evaporated using a high vacuum centrifuge . Samples were recovered by adding 500 μl of MeOH:H20 (75:25 V/V) and injected into HPLC for determining the total paclitaxel percentage content.
HPLC system
Column : Nova Pak C18 (Waters) 3.9 x 300 mm
Flow Rate 1.5 ml/m Detector UV 231 nm Injection 20 μl
Mobile Phase 57.5 % H20 brought to pH 2
42.5 % CH3CN
The present invention also provides a process for preparing a polymer conjugate which process comprises reacting a compound of the formula II
Figure imgf000007_0001
wherein one of A2 and A3 is a chemical bond and the other one is A, and A, R and R3 are as defined above, with an activated polymer consisting essentially of from 90 to 99.9 mol % of units represented by formula
I
CH2
I CH3 C-CO-NH-CH2-CHOH-CH3
and from 10 to 0.1 mol % of units represented by the formula .
Figure imgf000007_0002
wherein A, is as defined above and then τJreating the resultant polymer conjugate with 2-hydroxypropylamine. Preferably the reaction between the compounds of the formula II and the activated polymer is carried out in an anhydrous polar organic solvent such as dimethylsulfoxide or dimethylformamide optionally in presence of an organic or inorganic base such as an alkaline carbonate, dimethyla inopyridine or triethylamine. The reaction can typically be effected for from 1 to 24 hours.
The reaction is typically carried out at a temperature of from 15 to 40°C, preferably at room temperature. The alkaline carbonate is, for example, an alkali metal carbonate or an alkaline earth metal carbonate. Some of the starting compounds of the formula II are known compounds, i.e. paclitaxel or paclitaxel analogues or may be prepared starting from known compounds. For example 7-epi derivatives may be prepared by refluxing in toluene paclitaxel or its analogs in the presence of a base (Na2C03 or diazabicycloundecene) Other compounds of the formula II are new, in particular those in which A3 is βhla residue and those in which either A2 or A3 represents di, tri, or tetra peptide spacer as defined above for A and are within the scope of the invention.
The compounds of formula II wherein A2 is not a chemical single bond may be prepared by reacting a paclitaxel or a paclitaxel analog with protected amino acid or peptide in the presence of a condensating reagent, and with or without the additional presence of a catalyst, preferably at room temperature, followed by the removal of the protecting group with known methods. The condensation may be also carried out using activated esters such as paranitrophenyl ester of peptide or amino acid. Suitable condensing reagents include carbodiimides such as dicyclohexyl carbodii ide (DCC) . Suitable catalysts include 4-dimethylamino-pyridine (DMAP) , pyridine or triethylamine.
Various known amino protecting groups can be utilized and commercially available protected amino acid or peptideε can be utilized as the starting materials. Amino acids or peptides protected with t-BOC, trityl, FMOC or carbobenzyloxy (CBZ) can be utilized. Amino Acid or peptides procted with t-BOC, trityl or FMOC groups are preferred. The compounds of the formula II wherein A3 is not a chemical single bond may be prepared by protecting or blocking the 2'-hydroxy group and then esterifying the 7- position hydroxyl and then removing the 2'-protecting or blocking group. More preferably, the compounds of the formula II wherein A3 is Gly or Ala residue are prepared by reacting paclitaxel with 2-3 equivalents of N-protected amino acid to produce 2' ,7-disubstituted paclitaxel, the 2'-position amino acid is cleaved and then the 7-position amino acid is deprotected.
Reaction of paclitaxel and the protected amino acid is conducted in the presence of a condensing reagent and a catalyst, like those above defined. Cleavage of the 2'-amino acid is conducted by adjusting the pH of the 2'-7-(amino acid) paclitaxel solution to pH 7-7.4 for example by mixture of the 2' ,7-di(amino acid) paclitaxel in a phosphate buffer at pH 7-7.4 or with a slight excess of NaHC03. Deprotection of the amino acid is conducted under a known amino acid deprotection method, such as mild acid treatment with, for example, acetic acid, or by reduction. Thus, for example, paclitaxel is allowed to react with 2- 3 mol. equivalent of N-protected amino acid (t.boc, CBZ or FMOC protected) in methylene chloride in the presence of DCC and a catalytic amount of 4-dimethylaminopyridine. In this manner, the protected amino acid is introduced at 2' and 7-position. The 2',7-bis amino acid derivative of paclitaxel is allowed to stand in presence of NaHC03 in H20/MeOH for 2-5 hours, whereby selective deprotection at the 2'-position occurs to yield the 7-substituted derivative of paclitaxel. The protecting groups are removed by appropriate deprotecting agent (e.g., acid, mild base or hydrogenolysis) .
The activated polymer is a synthetic, water soluble polymer prepared by the copolymerization of N'-(2- hydroxypropyl) methacrylamide with p-nitrophenylesters of N-methacryloyl oligopeptides, as described in US-A- 4062831 and US-A-4097470.
The polymer conjugates of the formula I and the new paclitaxel derivatives of the formula II exhibit good water solubility, biocompatibility and release the paclitaxel or paclitaxel derivative in the plasma or after internalization into cells by cleaving of the oligopeptide spacers. Biological activity Microtubule assembly and disassembly assay
Calf brain tubulin was prepared by two cycles of assembly-disassembly (Shelanski M.L. Gaskin F. and Cantor C.R., Proc. Natl. Acad.Sci. U.S.A. 70, 765-768, 1973) and stored in liquid nitrogen in MAB (0.1 M MES, 2.5 mM EGTA, 0.5 mM MgS04, 0.1 mM EDTA, 0.1 mM DTT pH 6.4) . All the experiments were carried out on protein stored for less than 4 weeks. Before each experiment, the tubulin was kept 30 min at 4°C.
Assembly was monitored by the method of Gaskin et al (Gaskin F. , Cantor C.R. Shelanski M.L. J. Molec . Biol 89, 737-758, 1974). The cuvette (1 cm path) containing tubulin (1 mg/ml) and 1 mM GTP was shifted to 37°C and continous turbidity measurements were made at 340 nm on Perkin-Elmer 557 Double Wavelenght Double Beam Spectrophotometer equipped with an automatic recorder and a termostatically regulated sample chamber. After 30 minutes 4 mM CaCl2 was added and depolymerisation was measured for 10 minutes as decreased turbidity. At regular intervals of 15 minutes scalar doses of the tested compounds were added and variations in the turbidity were monitored. Data are espressed as percentage of ripolymerisation induced by the tested compounds. The results are shown in Table I. In vitro drug sensitivity assay
Exponentially growing B16-F10 murine melanoma cells were seeded (2 x 104/ml) in RPMI 1640 medium were supplemented with 10% heat-inactivated fetal calf serum and 2 mM glutamine in 24 well-plates (Costar) . Scalar concentrations of tested compounds were added immediately after seeding. The inhibition of cell growth was evaluated by counting cells with a coulter counter after 72 hrs incubation. For each tested compound concentration triplicate cultures were used. The antiproliferative activity of the tested compounds was calculated from dose-response curves and espressed as IC50 (dose causing 50% inhibition cell growth in treated cultures relative to untreated controls). The result are shown in Table I.
Table I
Figure imgf000012_0001
The copolymer-paclitaxel prepared in Example 6 was tested in vivo against B16 F10 murine melanoma in comparison with paclitaxel.
Mice C57B16 female mice were obtained from Charles River Italy.
Animals were 8 to 10 weeks old at the beginning of the experiment. Drugs Because of its limited aqueous solubility, paclitaxel was dissolved in a vehicle consisting of polyoxyethylated castor oil (Cremophor EL) 50% and ethanol 50% and then diluted with glucose 5% solution at the desidered concentrations. The solution was slightly hazy and precipitates formation was observed after short time.
The compound of example 6 was easily dissolved in glucose 5% and the resulting solution was clear for long time (more than 2 hours) . Final concentrations were referred to the paclitaxel content of the compound (4% of total) . Tumor
The murine melanoma B16F10 was used. A suspension of 105 tumor cells in 0.2 ml was injected εubcutaneously in the flank of mice. Tumor size was measured with caliper and tumor weight was calculated with the formula:
a ~ +b Drugs administration
Paclitaxel was given intraperitoneally because of its poor solubility and vehicle toxicity.
The compound of example 6 was injected intravenously. Both compounds were administered at day 1, 5, 9 after tumor implantation.
The data reported in Table 2 show that the compound of the present invention is more active than paclitaxel. The dose of the polymer-conjugate is referred to the paclitaxel content.
Table 2
Figure imgf000014_0001
TOX: number of mice which died for toxicity. TOX determination was made when mice died before the control or when significant body weight loss and/or spleen and or/or liver size reduction were observed. From the above data, it can be seen that the polymer conjugates of the present invention exhibit excellent antitumor activity. These compounds, therefore, are useful antitumor agents due to the lower toxicity and increased water solubility as compared to paclitaxel or its derivative. Examples of tumors that can be treated are for istance, sarcomas, carcinoma, lymphomas, neuroblastoma, melanoma, myeloma, Wilms tumor, leukemias and adenocarcinoma. The improved solubility ad decreased toxicity of the polymer-conjugates of the present invention means that they are suitable for intravenous injection or infusion. The dosage depends upon the age, weight and condition of the patient. The dosage may be from 1 mg/kg body weight to 1 g/kg body weight, preferably from 4 to 800 mg/kg body weight. Typical formulations contain a quantity of polymer-bound paclitaxel/poly erbound paclitaxel derivative equivalent to 0.5, 1.5, 10, 20, 25, or 50 mg of the active paclitaxel/paclitaxel derivative. The polymer conjugates may be formulated as pharmaceutical compositions with a pharmaceutically acceptable carrier or diluent. Any appropriate carrier or diluent may be used. The solutions for intravenous injection or infusion may contain as carrier or diluent, for example, sterile water or preferably they may be in the form of sterile, aqueous or isotonic saline solutions. The following examples illustrate the invention. Example 1
Copolymer of l-methacryloylamino-2-hydroxypropane, 1 ( ethacryloyl-glycyl-phenylalanyl-leucyl-glycyl)amino- 2-hydroxypropane and 2' (methacryloyl-glycyl-phenyl alanyl-leucyl-glycyl)paclitaxel.
To a solution of 1.4 g of copolymer of 1-methacryloyl- amino-2-hydroxypropane and N-(methylencarbonyl-Phe- Leu- Gly 4-nitrophenoxy)methacrylamide, prepared according to J. Kopecek et al., Makromol Chem 177. p. 2833 (1976) , in 15 ml of anhydrous dimethylformamide were added 100 mg of paclitaxel and 15 mg of dimethylaminopyridine. The yellow solution was stirred for 8 hours at room temperature under anhydrous conditions. Then 2-hydroxy propyla ine (0.2 ml) was dropped into reaction flask, and then the whole was stirred for 30 minutes.
The reaction solution was quenched with 0.3 ml glacial acetic acid, concentrated under vacuum to a small volume, and then poured into 200 ml of acetone. Ater 30' mixing, the precipitate was filtered and washed with acetone to yield 1.25 g of the title compound.
The paclitaxel content was 4.5% (evaluated by enzymatic hydrolysis and HPLC analysis) .
Unreacted paclitaxel was recovered from acetone solution. Example 2 2' (N-trityl-phenylalanyl-leucyl-glycyl)paclitaxel
To a solution of 170 mg of paclitaxel in 16 ml of acetonitrile were added 24 mg of dimethyla ino-pyridine and 150 mg of N-trityl-phenylanyl-leucyl-glycine 4- nitrophenyl ester.
The yellow solution was stirred for 20 hours at room temperature, and then evaporated under vacuum to dryness.
The residue was cromatographed on silica gel with ethyl- acetate-hexane 35:25 as eluant, affording 380 mg of the title compound.
Η-NMR(400 MHZ, CDC13) : δ
0.82 (d, J=6.4 Hz, 3H, S-Leu)
0.85 (d, J=6.7 Hz, 3H, S-Leu) 1.15 (ε, 3H, 16)
1.26 (s, 3H, 17)
1.2-1.6 (m. 3H, j3+j3+7-Leu)
1.69 (S, 3H, 19)
1.85 (s, 1H, OH-1) 1.89 (m, 1H, 6/3)
1.96 (d, J=1.2 HZ, 3H, 18)
2.14 (dd, J=5.9 Hz, J=13.5 Hz, 1H, 5-Phe)
2.24 (S, 3H, CH3CO-10)
2.2-2.7 (m, 5H, CH2-14+OH-7+6α+j8Phe-i-NH-Phe) 2.47 (s, 3H, CH3CO-4)
3.50 (m, 1H, α-Phe)
3.74 (dd, J=4.7 Hz, J=18.2 Hz, 1H, α-Gly)
3.80 ( , 1H, α-Leu)
3.83 (d, J=7.0 Hz, 1H, 3) 4.17 (dd, J=7.0 Hz, J=18.2 Hz, 1H, α'-Gly)
4.22, 4.33 (two-d, J=8.5 Hz, 2H, CH2-20)
4.46 (m, 1H, 7)
4.97 (dd, J=2.2 Hz, J=9.9 Hz, 1H, 5) 5.44 (d, J=2.3 HZ, 1H, 2') 5.71 (d, J=7.0 HZ, 1H, 2)
5.97 (dd, J=4.7 Hz, J=7.0 Hz, 1H, NH-Gly) 6.07 (dd, J=2.3 Hz, J=9.4 Hz, 1H, 3') 6.2-6.3 (m. 2H, 13+10)
6.8-9.2 (in, 30 H, 6- Ph) 6.95 (d, J=6.7 Hz, 1H, NH-Leu) 8.00 (d, J=9.4 HZ, 1H, NH-4' ) Example 3 2' (phenylalanyl-leucyl-glycyl)paclitaxel.
The compound 2' (N-Trit-Phe-Leu-Gly)paclitaxel (250 mg) was dissolved into a mixture of glacial acetic (22 ml) and water (6 ml) and the whole was stirred for 1 hour at room temperature. The solvents were evaporated in vacuum to the dryness, the residue stirred with diethylether-hexane 1:1 for 30 minutes and filtered to obtain 160 mg of the title compound.
FAB-MS: m/z 1171, M+H*] +; 1112, M-CH3C00H + 2H; 1051, 1024, 911, 603, 569, 509. Η-NMR(400MHz. CDC1, : δ 0.88(d,J=6.4Hz,3H, <5 Leu) 0.92(d,J=6.4Hz,3H, δ' Leu) 1.13 (s, 3H, 16) 1.16 (s, 3H, 17)
1.4-2.0(m,4H, + ' +yheu+ββ)
1.69 (s,3H, 19)
1.91 (d,J=1.2Hz,3H,18) 2.16 (dd, J=6.0 Hz,J=13.8Hz,lH,14)
2.23 (s, 3H, COCH3-10)
2.4-2.6 (m, 3H, 6α+14+0Phe)
2.53 (Ξ,3H,COCH3-4) 2.90 (dd, J=4.1Hz, J=13.5Hz, lH,β'-Phe)
3.49 (dd, J=4.1 Hz, J=9.1Hz,lH, Phe)
3.82 (d, J=7.3Hz,lH,3)
3.9-4.1 (m,2H, + 'Gly)
4.22, 4.33 (two-d, J=8.7 Hz , 2H, CH2-20) 4.27 (m, 1H, α-Ltυ)
4.44 (dd, J=6.4Hz, J=10.8Hz,7)
4.98 (dd, J=2.4Hz, J=9.7 Hz,5)
5.61 (d, J=3.2Hz,lH,2')
5.70 (d, J=7.3HZ,1H,2) 6.12 (dd,J=3.2Hz,J=9.4Hz,lH,3')
6.21 (m, 1H, 13)
6.28 (S,1H,10)
6.8-8.2 (m,21H,4-Ph + NHLeu)
7.87 (d,J=9.4Hz,lH,NH-4') Example 4
Copolymer of l-methacryloylamino-2-hydroxypropane, 1-
(methacry loy 1-glycy 1 ) amino-2 -hydroxypropane and
2 ' (methacryloyl-glycyl-phenylalanyl-leucyl- glycyl) paclitaxel. To a solution of 1 g of copolymer of 1-methacryloylamino-
2 -hydroxypropane and N- (methylencarbonyl-4-nitro phenoxy)methacrylamide, prepared according to P.
Rejmanova et al. , Makromol. Chem. 178 , p. 2159-2168, in 10 ml of anhydro dimethylformamide were added 100 mg of 2' Phe-Leu-Gly-paclitaxel and 10 mg of dimethylaminopyridine.
The yellow solution was stirred for 2 hours at room temperature under anhydrous conditions. Then 2-hydroxy - propylamine (0.15 ml) was added to the reaction solution, and the whole was stirred for 30 minutes. The reaction solution was quenched with 0.2 ml of glacial acetic acid, concentrated under vacuum to a small volume and then poured into 200 ml of acetone.
The mixture was stirred for 1 hours, the precipitate was filtered and washed with acetone to yield 960 mg of the title compound. The paclitaxel content was 6% (evaluated by enzymatic hydrolysis and HPLC analysis) . Example 5
Copolymer of l-methacryloylamino-2-hydroxy -propane, 1(methacryloyl-glycyl)amino-2-hydroxypropane and 2' (methacryloyl-glycyl)paclitaxel. To a solution of 1.6 g of copolymer of 1-methacryloyl - amino-2-hydroxypropane and N-(methylen-carbonyl-4-nitro- phenoxy)methacrylamide in 16 ml of anhydrous dimethyl¬ formamide were added 100 mg of paclitaxel and 20 mg of dimethylamino-pyridine. The yellow solution was stirred for 20 hours at room temperature, then 2- hydroxypropylamine (0.2 ml) was added, and the whole was stirred for 30 minutes. The reaction solution was quenched with 0.3 ml of glacial acetic acid, concentrated under vacuum to a small volume, and then poured into 200 ml of acetone. The mixture was stirred for 1 hour, the precipitate was filtered and washed with acetone to yield 1440 mg of the title compound. The paclitaxel content was 2.75% w/w% Example 6
Copolymer of 1-methacryloylamino -2-hydroxypropane, 1- (methacryloyl-glycyl-phenylanyl-leucyl-glycyl) amino-2- hydroxypropane and 2' (methacryloyl-glycyl-phenylalanyl- leucyl-glycyl-βalanyl)paclitaxel.
To a solution of 620 mg of copolymer of 1-metacrylamino- 2-hydroxypropane and N-(methylencarbonyl-Phe-Leu-Gly-4- nitro- phenoxy)methacrylamide in 6 ml of anhydrous dimethyl-formamide were added 62 mg of 2'- (βalanyl)paclitaxel, prepared according to N.F. Magri et al, J Nat. Products 5_1 298-306, 1988, and 10 mg of dimethylamino pyridine. The yellow solution was stirred for 5 hours at room temperature under anhydrous conditions. Then 2-hydroxypropylamine (0.1 ml) was added and the whole was stirred for 30 minutes.
The reaction solution was quenched with 0,15 ml of glacial acetic acid, concentrated under vacuum to a small volume and poured in 150 ml of acetone. The mixure was stirred for 1 hour, the precipitate was filtered and washed with acetone to yield 5.85 mg of the title compound. The paclitaxel content was 4% w/w% Example 7
2' ,7-di(carbobenzyloxy-/3-alanyl)paclitaxel To a solution of 200 mg of paclitaxel in 15 ml of acetonitrile were added 400 mg of N,N'- dicyclohexylcarbodiimide, 200 mg of carbobenzyloxy-β- alanine and 60 mg of dimethylamino-pyridine. The reaction mixture was stirred for 20 hours, the precipitate was filtered and the solvent evaporated under vacuum to dryness. The residue was chromatographed on silica gel with ethyl acetate-hexane 1:1 as eluant, affording 300 mg of the title compound. FAB-MS: m/z 1264 M+H] +, 1204, 1130, 1070
Example 8
7-(carbobenzyloxy-βalanyl)paclitaxel
To a solution of 171 mg of 2'7-di(carbobenzyloxy-/3- alanyl)paclitaxel in 60 ml of methanol, were added 30 mg of sodium bicarbonate and 7 ml of water . The reaction mixture was stirred for 3 hours at room temperature. The methanol was evaporated and the product extracted with ethylacetate.
The solvent was evaporated under vacuum to dryness, affording 134 mg of the title compound.
Example 9
7-(βalanyl)paclitaxel
To a solution of 135 mg of 7-(carbobenzyloxy- βalanyl)paclitaxel in 20 ml of methanol and 13 ml of formic acid, was added 200 mg of Pd/C 5%. The reaction mixture was stirred for 6 hours at room temperature. The catalyst was filtered, washed with methanol and the solvents were evaporated to dryness under vacuum. The residue was dissolved in 8 ml of methanol and precipitated with 150 ml of diethylether, affording 85 mg of the title compound.
FAB-MS: M/z 925,M+H]+ ; 947, M+Na~| +
*H NMR(400MHz, CDC13) : δ
1.14 (s,3H,CH3-16) 1.20(s,3H,CH3-17)
1.79 (Ξ,3H,CH3-19)
1.85 (s, 3H,CH3-18) 2.17(s,3H,CH3CO-10)
2.2-2.6 (m, 6H,CH2-14+CH2-6+OCOCH2CH2NH2) 2.42(s,3H,CH3CO-4)
3.0-3.2 (i, 2H ,OCOCHCHoNH,) 3.90(d,J=6.8Hz,lH,3)
4.18, 4.31(two d, J=8.2Hz,2H,CH2-20)
4.80(d,J=3.2Hz,lH,2')
4.91(d,J=8.5Hz,lH,5)
5.62(dd,J=10.2Hz, J=7.0Hz, 1H,7)
5.66 (d,J=6.8Hz,lH,2)
5.81(dd,J=2.9Hz, J=9.1Hz, 1H, 3 '
6.17 (m,lH,13)
6.19(s,lH,10)
7.3-8.2 (m,16H,NH-4'+3-Ph)
Example 10
Copolymer of l-methacryloylamino-2-hydroxypropane, 1- (methacryloyl-glycyl-phenylalanyl-leucyl-glycyl)amino-2- hyroxypropane and 7-(methacryloyl-glycyl-phenylalanyl- leucyl-glycyl-jSalanyl)paclitaxel. To a solution of 1500 mg of copolymer of 1-methacryloyl- amino-2-hydroxypropane and N-(methylencarbonyl-Phe-Leu- Gly-4nitrophenoxy)methacrylamide in 13 ml of anhydrous dimethylformamide were added 135 mg of 7- (βalanyl)paclitaxel and 20 mg of dimethylaminopyridine. The yellow solution was stirred under anhydrous conditions for 5 hours at room temperature. Then 2-hydroxypropylamine (0.2 ml) was added , and the whole was stirred for 30 minutes.
The reaction solution was quenched with 0.3 ml of glacial acetic acid, concentrated under vacuum to a small volume and poured in 250 ml of acetone. The mixture was stirred for 1 hour, the precipitate was filtered and washed with acetone to yield 1520 mg of the title compound. The paclitaxel content was 7.8% w/w%.

Claims

A polymer conjugate consisting essentially of: from 90 to 99.9 mol % of units represented by the formula
CH,
CH3—C-CO-NH-CH2-CHOH-CH3 I
from 0.1 to 5 mol % of units represented by the formula
Figure imgf000026_0001
wherein one of R, and R2 is a copolymer residue of the formula
CH,
CH, C-CONH-CH,-CO-A—
and the other one is hydrogen atom; from 0 to 9.9 mol % of units represented by the formula I
CH2
I CH3 — C-CONH-CH2-CO-A,-NH-CH2-CHOH-CH3
wherein R is a phenyl or t-butoxy group, R3 is H or an acetyl group, A and A, which may be the same or different, represent a chemical single bond, an amino acid residue or peptide spacer selected from β Ala, Gly, Phe-Gly, Phe-Phe-, Leu-Gly, Val-Ala, Phe-Ala, Leu- Phe, Leu-Ala, Phe-Leu-Gly, Phe-Pl.e-Leu, Leu-Leu-Gly, Phe- Tyr-Ala, Phe-Gly-Phe, Phe-Phe-Gly, Phe-Leu-Gly-Phe, Gly- Phe-Leu-Gly-Phe,Gly- Ala, Phe-Gly-βAla, Phe-Phe-βAla, Leu-Gly-βAla, Val-Ala-βAla, Phe-Ala-βAla, Leu-Phe-βAla, Leu-Gly-βAla, Phe-Leu-Gly-βAla, Phe-Phe-Leu βAla, Leu- Leu-Gly-βAla, Phe-Tyr-Ala-βAla, Phe-Gly-Phe-β, Phe-Phe- Gly βAla, Phe-Leu-Gly-Phe-βAla or Gly-Phe-Leu-Gly-Phe- jSAla.
2. A polymer conjugate according to claim 1, in which R represents a phenyl group, R3 is an acetyl group and A is a Phe-Leu-Gly or Phe-Leu-Gly-βAla residue.
3. A polymer conjugate according claim 1 or 2 , which is a copolymer of l-methacryloylamino-2-hydroxypropane,
(methacryloylglycyl-phenylalanylleucylglycyl) 3-amino-2 hydroxypropane and 2 '-(methacryloylglycylphenylalanyl- leucylglycyl-βalanyl) paclitaxel.
4. A process for preparing a polymer conjugate as defined in claim 1, which process comprises reacting a compound of the formula II
Figure imgf000028_0001
wherein one of A2 and A3 is a chemical bond and the other one is A, and A, R and R3 are as defined in claim 1, with an activated polymer consisting essentially of from 90 to 99.9 mol % of units represented by formula
I
CH2
I
CH3 C-CO-NH-CH2-CHOH-CH3
I and from 10 to 0.1 mol % of units represented by the formula ■
CH,
CH ,—C-CO-NH-CH2-CO-Aι-0- Q ~N02 wherein A, is as defined in claim 1 and then treating the resultant polymer conjugate with 2-hydroxy- propylamine.
5. A process according to claim 4, in which the reaction is carried out in an anhydrous polar solvent at a temperature of from 15 to 40°C for from 1 to 24 hours, in the presence of an organic or inorganic base.
6. A compound of the formula II as defined in claim 4 wherein A2 represents di, tri or tetra peptide spacer as defined in claim 1 for A.
7. A process for preparing a compound of the formula II as defined in claim 6, which process comprises reacting a paclitaxel or a paclitaxel analog with desired protected amino acid or peptide in the presence of a condensating reagent or using activated esters and with or without the additional presence of a catalyst and then removing the protecting groups.
8. A compound of formula II as defined in claim 4 wherein A3 represents βAla or di, tri or tetra peptide spacer as defined in claim 1 for A.
9. A process for preparing a compound of the formula II as defined in claim 8, which process comprises either (i) protecting or blocking the 2'-hydroxy group of paclitaxel and esterifying the 7-position hydroxyl and then removing the 2'-protecting or blocking group, or
(ii) reacting paclitaxel with 2-3 moles of the desired N- protected amino acid to produce 2' ,7-disubstituted taxol, cleaving the 2'-position amino acid and then deprotecting the 7-position amino acid.
10. A pharmaceutical composition comprising, as active ingredient, a polymer conjugate as defined in claim 1, 6 or 8 and a pharmaceutically acceptable diluent or carrier.
PCT/EP1993/001433 1992-06-19 1993-06-07 Polymer-bound paclitaxel derivatives WO1994000156A1 (en)

Priority Applications (12)

Application Number Priority Date Filing Date Title
AT93912905T ATE205725T1 (en) 1992-06-19 1993-06-07 POLYMER BONDED PACLITAXEL DERIVATIVES
KR1019940700488A KR100281606B1 (en) 1992-06-19 1993-06-07 Polymer-linked paclitaxel derivatives, methods for their preparation and pharmaceutical compositions comprising the same
PL93302437A PL173898B1 (en) 1992-06-19 1993-06-07 Paklitaxel derivatives bound with a polymer
DE69330776T DE69330776T2 (en) 1992-06-19 1993-06-07 Polymer-bound paclitaxel derivatives
RU94016158A RU2130462C1 (en) 1992-06-19 1993-06-07 Polymeric conjugate, a method of its synthesis and a pharmaceutical composition
EP93912905A EP0600062B1 (en) 1992-06-19 1993-06-07 Polymer-bound paclitaxel derivatives
AU43233/93A AU659750B2 (en) 1992-06-19 1993-06-07 Polymer-bound paclitaxel derivatives
CZ94620A CZ62094A3 (en) 1992-06-19 1993-06-07 Paclitaxel derivatives bound onto a polymer
JP50198194A JP3693340B2 (en) 1992-06-19 1993-06-07 Polymer-bound paclitaxel derivatives
UA94005444A UA39926C2 (en) 1992-06-19 1993-07-06 POLYMER CONJUGATE, PHARMACEUTICAL COMPOSITION WITH ANTI-TUMOR ACTIVITY
FI940733A FI940733A0 (en) 1992-06-19 1994-02-16 Polymer-linked paclitaxel derivatives
NO940567A NO940567L (en) 1992-06-19 1994-02-18 Polymer-bound paclitaxel derivatives

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
GB929213077A GB9213077D0 (en) 1992-06-19 1992-06-19 Polymerbound taxol derivatives
GB9213077.2 1992-06-19

Publications (1)

Publication Number Publication Date
WO1994000156A1 true WO1994000156A1 (en) 1994-01-06

Family

ID=10717411

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP1993/001433 WO1994000156A1 (en) 1992-06-19 1993-06-07 Polymer-bound paclitaxel derivatives

Country Status (25)

Country Link
US (4) US5362831A (en)
EP (1) EP0600062B1 (en)
JP (1) JP3693340B2 (en)
KR (1) KR100281606B1 (en)
CN (1) CN1041281C (en)
AT (1) ATE205725T1 (en)
AU (2) AU659750B2 (en)
CA (1) CA2112482A1 (en)
CZ (1) CZ62094A3 (en)
DE (1) DE69330776T2 (en)
ES (1) ES2164663T3 (en)
FI (1) FI940733A0 (en)
GB (1) GB9213077D0 (en)
HU (2) HUT67914A (en)
IL (1) IL106023A (en)
MX (1) MX9303598A (en)
MY (1) MY109213A (en)
NZ (1) NZ253116A (en)
PL (1) PL173898B1 (en)
RU (1) RU2130462C1 (en)
SG (1) SG49248A1 (en)
TW (1) TW266201B (en)
UA (1) UA39926C2 (en)
WO (1) WO1994000156A1 (en)
ZA (1) ZA934388B (en)

Cited By (27)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1995010304A1 (en) * 1993-10-08 1995-04-20 Pharmacia S.P.A Polymer-bound camptothecin derivatives
EP0693485A3 (en) * 1994-06-28 1996-05-08 Tanabe Seiyaku Co Baccatin derivatives and processes for preparing the same
US5608073A (en) * 1994-11-17 1997-03-04 Tanabe Seiyaku Co., Ltd. Baccatin derivatives and processes for preparing the same
EP0807115A1 (en) * 1995-01-30 1997-11-19 Enzon, Inc. High molecular weight polymer-based prodrugs
WO1998034968A1 (en) * 1997-02-11 1998-08-13 The Council Of The Queensland Institute Of Medical Research Polymers incorporating peptides
US5886026A (en) * 1993-07-19 1999-03-23 Angiotech Pharmaceuticals Inc. Anti-angiogenic compositions and methods of use
EP0923566A1 (en) * 1996-08-20 1999-06-23 Enzon, Inc. High molecular weight polymer-based prodrugs
EP0932399A1 (en) * 1996-03-12 1999-08-04 PG-TXL Company, L.P. Water soluble paclitaxel prodrugs
EP1251739A2 (en) * 1999-04-13 2002-10-30 Fannin Bioscience, Inc. Poly(dipeptide) as a drug carrier
EP2014307A2 (en) 2001-03-13 2009-01-14 Angiotech International Ag Micellar drug delivery vehicles and uses thereof
EP2108390A2 (en) 2008-03-31 2009-10-14 Cordis Corporation Device for local and/or regional delivery employing liquid formulations of therapeutic agents
EP2123312A2 (en) 2008-05-19 2009-11-25 Cordis Corporation Extraction of solvents from drug containing polymer reservoirs
EP2181704A2 (en) 2002-12-30 2010-05-05 Angiotech International Ag Drug delivery from rapid gelling polymer composition
US7846940B2 (en) 2004-03-31 2010-12-07 Cordis Corporation Solution formulations of sirolimus and its analogs for CAD treatment
US7875677B2 (en) 2001-04-20 2011-01-25 The University Of British Columbia Micellar drug delivery systems for hydrophobic drugs
AU2007254682B2 (en) * 1993-07-19 2011-06-02 Angiotech Pharmaceuticals, Inc. Anti-angiogenic compositions and methods of use
US7989490B2 (en) 2004-06-02 2011-08-02 Cordis Corporation Injectable formulations of taxanes for cad treatment
EP2380606A1 (en) 2006-06-30 2011-10-26 Cook Incorporated Methods of manufacturing and modifying taxane coatings for implantable medical devices
US8313521B2 (en) 1995-06-07 2012-11-20 Cook Medical Technologies Llc Method of delivering an implantable medical device with a bioabsorbable coating
WO2012162007A1 (en) 2011-05-25 2012-11-29 Cordis Corporation Expandable devices coated with a rapamycin composition
WO2012162010A1 (en) 2011-05-25 2012-11-29 Cordis Corporation Expandable devices coated with a paclitaxel composition
US8409601B2 (en) 2008-03-31 2013-04-02 Cordis Corporation Rapamycin coated expandable devices
US8420110B2 (en) 2008-03-31 2013-04-16 Cordis Corporation Drug coated expandable devices
WO2014004760A1 (en) 2012-06-28 2014-01-03 Covidien Lp Post-processing of a medical device to control morphology and mechanical properties
US8642063B2 (en) 2008-08-22 2014-02-04 Cook Medical Technologies Llc Implantable medical device coatings with biodegradable elastomer and releasable taxane agent
US9085605B2 (en) 2010-05-27 2015-07-21 Shenzhen Salubris Pharmaceuticals Co., Ltd. Chemical synthesis and anti-tumor and anti-metastatic effects of dual functional conjugate
WO2017053920A1 (en) 2015-09-25 2017-03-30 Zy Therapeutics Inc. Drug formulation based on particulates comprising polysaccharide-vitamin conjugate

Families Citing this family (126)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE69118826T2 (en) * 1990-11-27 1996-11-14 Fuji Photo Film Co Ltd Propenamide derivatives, their polymers, copolymers and their use
GB9213077D0 (en) * 1992-06-19 1992-08-05 Erba Carlo Spa Polymerbound taxol derivatives
US5614549A (en) * 1992-08-21 1997-03-25 Enzon, Inc. High molecular weight polymer-based prodrugs
WO1995003036A1 (en) 1993-07-19 1995-02-02 Angiogenesis Technologies, Inc. Anti-angiogenic compositions and methods of use
US20030203976A1 (en) 1993-07-19 2003-10-30 William L. Hunter Anti-angiogenic compositions and methods of use
JPH09504033A (en) * 1993-10-20 1997-04-22 エンゾン,インコーポレーテッド 2'- and / or 7-substituted taxoids
US5580898A (en) * 1994-05-24 1996-12-03 The Trustees Of The University Of Pennsylvania Method of stabilizing microtubules
US5626862A (en) 1994-08-02 1997-05-06 Massachusetts Institute Of Technology Controlled local delivery of chemotherapeutic agents for treating solid tumors
US5801191A (en) * 1995-06-01 1998-09-01 Biophysica Foundation Taxoids
US5840748A (en) * 1995-10-02 1998-11-24 Xechem International, Inc. Dihalocephalomannine and methods of use therefor
US5807888A (en) * 1995-12-13 1998-09-15 Xechem International, Inc. Preparation of brominated paclitaxel analogues and their use as effective antitumor agents
US5854278A (en) * 1995-12-13 1998-12-29 Xechem International, Inc. Preparation of chlorinated paclitaxel analogues and use thereof as antitumor agents
US5654448A (en) * 1995-10-02 1997-08-05 Xechem International, Inc. Isolation and purification of paclitaxel from organic matter containing paclitaxel, cephalomannine and other related taxanes
US6177456B1 (en) 1995-10-02 2001-01-23 Xechem International, Inc. Monohalocephalomannines having anticancer and antileukemic activity and method of preparation therefor
US6441025B2 (en) * 1996-03-12 2002-08-27 Pg-Txl Company, L.P. Water soluble paclitaxel derivatives
EP0895784B1 (en) 1996-04-15 2005-11-23 Asahi Kasei Kabushiki Kaisha Drug complexes comprising taxane compounds or steroids
US5919815A (en) * 1996-05-22 1999-07-06 Neuromedica, Inc. Taxane compounds and compositions
EP0914116B1 (en) * 1996-05-22 2000-10-11 Protarga Inc. Compositions comprising conjugates of cis-docosahexaenoic acid and taxotere
US5795909A (en) 1996-05-22 1998-08-18 Neuromedica, Inc. DHA-pharmaceutical agent conjugates of taxanes
US6576636B2 (en) 1996-05-22 2003-06-10 Protarga, Inc. Method of treating a liver disorder with fatty acid-antiviral agent conjugates
DK0914102T3 (en) 1996-05-24 2006-01-09 Angiotech Pharm Inc Preparations and methods for treating or preventing diseases of the body canals
WO1998047496A2 (en) * 1997-04-18 1998-10-29 Access Pharmaceuticals, Inc. Polymer-platinum compounds
US5965118A (en) * 1997-04-18 1999-10-12 Access Pharmaceuticals, Inc. Polymer-platinum compounds
US8853260B2 (en) 1997-06-27 2014-10-07 Abraxis Bioscience, Llc Formulations of pharmacological agents, methods for the preparation thereof and methods for the use thereof
US6501188B1 (en) * 1997-07-03 2002-12-31 Micron Technology, Inc. Method for improving a stepper signal in a planarized surface over alignment topography
GB9721070D0 (en) 1997-10-03 1997-12-03 Pharmacia & Upjohn Spa Bioactive derivatives of camptothecin
GB9721069D0 (en) 1997-10-03 1997-12-03 Pharmacia & Upjohn Spa Polymeric derivatives of camptothecin
WO1999030727A1 (en) 1997-12-17 1999-06-24 Enzon, Inc. Polymeric prodrugs of amino- and hydroxyl-containing bioactive agents
US7208011B2 (en) 2001-08-20 2007-04-24 Conor Medsystems, Inc. Implantable medical device with drug filled holes
US20040254635A1 (en) 1998-03-30 2004-12-16 Shanley John F. Expandable medical device for delivery of beneficial agent
US6241762B1 (en) 1998-03-30 2001-06-05 Conor Medsystems, Inc. Expandable medical device with ductile hinges
US7208010B2 (en) 2000-10-16 2007-04-24 Conor Medsystems, Inc. Expandable medical device for delivery of beneficial agent
US6153655A (en) 1998-04-17 2000-11-28 Enzon, Inc. Terminally-branched polymeric linkers and polymeric conjugates containing the same
US6293967B1 (en) 1998-10-29 2001-09-25 Conor Medsystems, Inc. Expandable medical device with ductile hinges
US7018654B2 (en) 1999-03-05 2006-03-28 New River Pharmaceuticals Inc. Pharmaceutical composition containing an active agent in an amino acid copolymer structure
US7235583B1 (en) 1999-03-09 2007-06-26 Luitpold Pharmaceuticals, Inc., Fatty acid-anticancer conjugates and uses thereof
US7060708B2 (en) * 1999-03-10 2006-06-13 New River Pharmaceuticals Inc. Active agent delivery systems and methods for protecting and administering active agents
US6716452B1 (en) 2000-08-22 2004-04-06 New River Pharmaceuticals Inc. Active agent delivery systems and methods for protecting and administering active agents
US6290673B1 (en) 1999-05-20 2001-09-18 Conor Medsystems, Inc. Expandable medical device delivery system and method
US6669951B2 (en) 1999-08-24 2003-12-30 Cellgate, Inc. Compositions and methods for enhancing drug delivery across and into epithelial tissues
WO2001013957A2 (en) 1999-08-24 2001-03-01 Cellgate, Inc. Enhancing drug delivery across and into epithelial tissues using oligo arginine moieties
US6730293B1 (en) 1999-08-24 2004-05-04 Cellgate, Inc. Compositions and methods for treating inflammatory diseases of the skin
US7229961B2 (en) 1999-08-24 2007-06-12 Cellgate, Inc. Compositions and methods for enhancing drug delivery across and into ocular tissues
CN1111166C (en) * 1999-09-10 2003-06-11 云南汉德生物技术有限公司 Water soluble cephalotaxin phosphorus poly aminoacid ester or its salt, pharmaceutical compositions contg. same, and pharmaceutical use thereof
US20030054977A1 (en) * 1999-10-12 2003-03-20 Cell Therapeutics, Inc. Manufacture of polyglutamate-therapeutic agent conjugates
AU1820400A (en) * 1999-11-17 2001-05-30 School Of Pharmacy, University Of London, The Conjugates of hpma copolymer and ellipticin
US6452024B1 (en) 2000-02-22 2002-09-17 Chaichem Pharmaceuticals International Process for extraction and purification of paclitaxel from natural sources
US20020077290A1 (en) * 2000-03-17 2002-06-20 Rama Bhatt Polyglutamic acid-camptothecin conjugates and methods of preparation
US7163918B2 (en) 2000-08-22 2007-01-16 New River Pharmaceuticals Inc. Iodothyronine compositions
US20020099013A1 (en) * 2000-11-14 2002-07-25 Thomas Piccariello Active agent delivery systems and methods for protecting and administering active agents
WO2005032474A2 (en) * 2003-09-30 2005-04-14 New River Pharmaceuticals Inc. Pharmaceutical compositions for prevention of overdose or abuse
US6764507B2 (en) 2000-10-16 2004-07-20 Conor Medsystems, Inc. Expandable medical device with improved spatial distribution
DE20122506U1 (en) 2000-10-16 2005-12-08 Conor Medsystems, Inc., Menlo Park Expandable medical device for delivering a beneficial agent
US8394813B2 (en) * 2000-11-14 2013-03-12 Shire Llc Active agent delivery systems and methods for protecting and administering active agents
US6566556B2 (en) * 2000-12-19 2003-05-20 Nippon Shokubai Co., Ltd. Method for production of alkanolamine and apparatus therefor
US6964680B2 (en) 2001-02-05 2005-11-15 Conor Medsystems, Inc. Expandable medical device with tapered hinge
US20040073294A1 (en) 2002-09-20 2004-04-15 Conor Medsystems, Inc. Method and apparatus for loading a beneficial agent into an expandable medical device
EP1362053B1 (en) * 2001-02-20 2007-11-14 Enzon, Inc. Terminally-branched polymeric linkers and polymeric conjugates containing the same
FI114710B (en) * 2001-03-12 2004-12-15 Ctt Cancer Targeting Tech Oy Novel peptide ligands for leukocyte integrins
US20030157170A1 (en) * 2001-03-13 2003-08-21 Richard Liggins Micellar drug delivery vehicles and precursors thereto and uses thereof
US8552054B2 (en) 2001-03-23 2013-10-08 Luitpold Pharmaceuticals, Inc. Fatty amine drug conjugates
JP4634694B2 (en) 2001-03-23 2011-02-16 ルイトポルド・ファーマシューティカルズ・インコーポレーテッド Fatty alcohol drug complex
US7056338B2 (en) 2003-03-28 2006-06-06 Conor Medsystems, Inc. Therapeutic agent delivery device with controlled therapeutic agent release rates
US7338939B2 (en) * 2003-09-30 2008-03-04 New River Pharmaceuticals Inc. Abuse-resistant hydrocodone compounds
US7375082B2 (en) * 2002-02-22 2008-05-20 Shire Llc Abuse-resistant hydrocodone compounds
US20070066537A1 (en) * 2002-02-22 2007-03-22 New River Pharmaceuticals Inc. Compounds and compositions for prevention of overdose of oxycodone
US20060014697A1 (en) 2001-08-22 2006-01-19 Travis Mickle Pharmaceutical compositions for prevention of overdose or abuse
US7169752B2 (en) * 2003-09-30 2007-01-30 New River Pharmaceuticals Inc. Compounds and compositions for prevention of overdose of oxycodone
US7141540B2 (en) * 2001-11-30 2006-11-28 Genta Salus Llc Cyclodextrin grafted biocompatible amphilphilic polymer and methods of preparation and use thereof
US7659253B2 (en) 2002-02-22 2010-02-09 Shire Llc Abuse-resistant amphetamine prodrugs
US7105486B2 (en) * 2002-02-22 2006-09-12 New River Pharmaceuticals Inc. Abuse-resistant amphetamine compounds
US7700561B2 (en) * 2002-02-22 2010-04-20 Shire Llc Abuse-resistant amphetamine prodrugs
ES2500117T3 (en) * 2002-02-22 2014-09-30 Shire Llc Novel sustained release pharmaceutical compounds to prevent the abuse of controlled substances
IL163668A0 (en) * 2002-02-22 2005-12-18 New River Pharmaceuticals Inc Use of peptide-drug conjugation to reduce inter-subject variability ofdrug serum levels
CA2483696A1 (en) * 2002-05-06 2003-11-20 University Of Utah Research Foundation Preblocking with non-ha gags increases effectiveness of ha conjugated anticancer agents
ATE424187T1 (en) * 2002-07-15 2009-03-15 Alcon Inc BIOERODIBLE FILM FOR DELIVERING AN OPHTHALMIC MEDICINAL PRODUCT
EP2260874A1 (en) * 2003-01-06 2010-12-15 Angiochem Inc. Aprotinin analogs as carriers across the blood-brain barrier
US6759539B1 (en) 2003-02-27 2004-07-06 Chaichem Pharmaceuticals International Process for isolation and purification of paclitaxel from natural sources
EP1610823B1 (en) 2003-03-28 2011-09-28 Innovational Holdings, LLC Implantable medical device with continuous agent concentration gradient
EP1644019B2 (en) 2003-05-29 2018-02-21 Shire LLC Abuse resistant amphetamine compounds
US7169179B2 (en) 2003-06-05 2007-01-30 Conor Medsystems, Inc. Drug delivery device and method for bi-directional drug delivery
WO2005007819A2 (en) 2003-07-09 2005-01-27 Wisconsin Alumni Research Foundation Charge-dynamic polymers and delivery of anionic compounds
US7785653B2 (en) 2003-09-22 2010-08-31 Innovational Holdings Llc Method and apparatus for loading a beneficial agent into an expandable medical device
WO2005068460A1 (en) 2003-12-22 2005-07-28 Schering Corporation Isothiazole dioxides as cxc- and cc- chemokine receptor ligands
EP2433653B1 (en) 2005-07-15 2019-06-05 Angiochem Inc. Use of aprotinin polypeptides as carriers in pharmaceutical conjugates
US20070073385A1 (en) * 2005-09-20 2007-03-29 Cook Incorporated Eluting, implantable medical device
PT1966151E (en) 2005-12-13 2011-12-20 Schering Corp Polycyclic indazole derivatives that are erk inhibitors
EP2132178B1 (en) 2006-12-20 2015-08-19 Merck Sharp & Dohme Corp. Jnk inhibitors
CA2674378A1 (en) * 2007-01-03 2008-07-17 Burnham Institute For Medical Research Methods and compositions related to clot binding compounds
NZ588816A (en) 2007-01-21 2011-11-25 Hemoteq Ag Medical device for the treatment of stenoses of corporal lumina and for the prevention of impending restenoses
US8784866B2 (en) * 2007-03-26 2014-07-22 William Marsh Rice University Water-soluble carbon nanotube compositions for drug delivery and medicinal applications
US20080241215A1 (en) * 2007-03-28 2008-10-02 Robert Falotico Local vascular delivery of probucol alone or in combination with sirolimus to treat restenosis, vulnerable plaque, aaa and stroke
US9365634B2 (en) * 2007-05-29 2016-06-14 Angiochem Inc. Aprotinin-like polypeptides for delivering agents conjugated thereto to tissues
PE20090326A1 (en) 2007-06-05 2009-04-04 Schering Corp HETEROCYCLES COMPOUNDS AS ERK INHIBITORS
US9192697B2 (en) 2007-07-03 2015-11-24 Hemoteq Ag Balloon catheter for treating stenosis of body passages and for preventing threatening restenosis
WO2009070380A2 (en) * 2007-10-03 2009-06-04 William Marsh Rice University Water-soluble carbon nanotube compositions for drug delivery and medical applications
WO2009049100A2 (en) * 2007-10-09 2009-04-16 Wisconsin Alumni Research Foundation Ultrathin multilayered films for controlled release of anionic reagents
US8710013B2 (en) 2008-04-18 2014-04-29 Angiochem Inc. Pharmaceutical compositions of paclitaxel, paclitaxel analogs or paclitaxel conjugates and related methods of preparation and use
US9198968B2 (en) 2008-09-15 2015-12-01 The Spectranetics Corporation Local delivery of water-soluble or water-insoluble therapeutic agents to the surface of body lumens
WO2010043049A1 (en) 2008-10-15 2010-04-22 Angiochem Inc. Etoposide and doxorubicin conjugates for drug delivery
BRPI0920209A2 (en) 2008-10-15 2015-12-22 Angiochem Inc conjugates of glp-1 agonists and their uses
CA2745524C (en) 2008-12-05 2020-06-09 Angiochem Inc. Conjugates of neurotensin or neurotensin analogs and uses thereof
US8853353B2 (en) 2008-12-17 2014-10-07 Angiochem, Inc. Membrane type-1 matrix metalloprotein inhibitors and uses thereof
CZ303072B6 (en) * 2009-02-13 2012-03-21 Zentiva, K.S. Paclitaxel and docetaxel polymeric conjugates with pH controlled release of cancerostatic
MX2011011023A (en) 2009-04-20 2012-01-20 Angiochem Inc Treatment of ovarian cancer using an anticancer agent conjugated to an angiopep-2 analog.
EP2421571A2 (en) * 2009-04-24 2012-02-29 Boston Scientific Scimed, Inc. Use of drug polymorphs to achieve controlled drug delivery from a coated medical device
BRPI1015918A2 (en) 2009-07-02 2019-09-24 Angiochem Inc multimeric peptide conjugates and uses thereof
WO2011005421A2 (en) * 2009-07-10 2011-01-13 Boston Scientific Scimed, Inc. Use of nanocrystals for a drug delivery balloon
EP2453938B1 (en) 2009-07-17 2015-08-19 Boston Scientific Scimed, Inc. Nucleation of drug delivery balloons to provide improved crystal size and density
EP2483263B1 (en) 2009-09-30 2018-07-18 Merck Sharp & Dohme Corp. Heterocyclic compounds that are erk inhibitors
EP2611476B1 (en) 2010-09-02 2016-08-10 Boston Scientific Scimed, Inc. Coating process for drug delivery balloons using heat-induced rewrap memory
WO2013022458A1 (en) 2011-08-05 2013-02-14 Boston Scientific Scimed, Inc. Methods of converting amorphous drug substance into crystalline form
WO2013028208A1 (en) 2011-08-25 2013-02-28 Boston Scientific Scimed, Inc. Medical device with crystalline drug coating
RU2493848C1 (en) * 2012-06-14 2013-09-27 Геннадий Петрович Власов Biodegradable polymer carrier for anti-cancer drug delivery (versions)
AR099812A1 (en) 2014-03-21 2016-08-17 Abbvie Inc ANTI-EGFR ANTIBODY AND DRUG ANTIBODIES AND CONJUGATES
CN107921143B (en) 2015-06-15 2021-11-19 安吉奥开米公司 Method for treating leptomeningeal carcinomatosis
WO2017128173A1 (en) * 2016-01-28 2017-08-03 北京和理咨询有限公司 Aptamer conjugate of taxol or derivative thereof, and preparation method and application of same
BR112018075653A2 (en) 2016-06-08 2019-08-27 Abbvie Inc anti-b7-h3 antibodies and drug antibody conjugates
JP2019521973A (en) 2016-06-08 2019-08-08 アッヴィ・インコーポレイテッド Anti-BH7-H3 antibody and antibody drug conjugate
US20200147235A1 (en) 2016-06-08 2020-05-14 Abbvie Inc. Anti-cd98 antibodies and antibody drug conjugates
BR112018075626A2 (en) 2016-06-08 2019-03-19 Abbvie Inc. anti-b7-h3 antibodies and antibody drug conjugates
MX2018015274A (en) 2016-06-08 2019-10-07 Abbvie Inc Anti-cd98 antibodies and antibody drug conjugates.
US20200353090A1 (en) * 2017-11-17 2020-11-12 Centre National De La Recherche Scientifique Polymeric prodrugs and subcutaneous and/or intramuscular administration thereof
WO2021194298A1 (en) * 2020-03-26 2021-09-30 (주) 테라베스트 Nanoparticles comprising drug dimers, and use thereof
AR124681A1 (en) 2021-01-20 2023-04-26 Abbvie Inc ANTI-EGFR ANTIBODY-DRUG CONJUGATES
WO2023154464A1 (en) * 2022-02-11 2023-08-17 Fairleigh Dickinson University Locally administered compositions and methods of use thereof

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4097470A (en) * 1974-04-25 1978-06-27 Ceskoslovenska Akademie Ved Preparation of biogically active substances bearing -NH2 groups in a form releasable by enzymatic cleavage
EP0187547A2 (en) * 1985-01-04 1986-07-16 Ceskoslovenska akademie ved Synthetic polymeric drugs
EP0393575A1 (en) * 1989-04-17 1990-10-24 G.D. Searle & Co. Neoplasia treatment compositions containing antineoplastic agent and side-effect reducing protective agent
EP0537905A1 (en) * 1991-10-15 1993-04-21 Virginia Tech Intellectual Properties, Inc. Water soluble derivatives of taxol with anti-neoplastic activity

Family Cites Families (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CS173846B1 (en) * 1974-04-23 1977-03-31
US4690790A (en) * 1985-07-23 1987-09-01 Stemcor Corporation Silicon nitride/silicon carbide composition and articles thereof
US4960790A (en) * 1989-03-09 1990-10-02 University Of Kansas Derivatives of taxol, pharmaceutical compositions thereof and methods for the preparation thereof
US5200534A (en) * 1992-03-13 1993-04-06 University Of Florida Process for the preparation of taxol and 10-deacetyltaxol
DE69325454T2 (en) * 1992-04-17 2000-01-20 Abbott Lab TAXOL DERIVATIVES
GB9213077D0 (en) * 1992-06-19 1992-08-05 Erba Carlo Spa Polymerbound taxol derivatives

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4097470A (en) * 1974-04-25 1978-06-27 Ceskoslovenska Akademie Ved Preparation of biogically active substances bearing -NH2 groups in a form releasable by enzymatic cleavage
EP0187547A2 (en) * 1985-01-04 1986-07-16 Ceskoslovenska akademie ved Synthetic polymeric drugs
EP0393575A1 (en) * 1989-04-17 1990-10-24 G.D. Searle & Co. Neoplasia treatment compositions containing antineoplastic agent and side-effect reducing protective agent
EP0537905A1 (en) * 1991-10-15 1993-04-21 Virginia Tech Intellectual Properties, Inc. Water soluble derivatives of taxol with anti-neoplastic activity

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
J. MED. CHEM. vol. 35, 1992, pages 145 - 151 A. E. MATHEW ET AL. 'SYNTHESIS AND EVALUATION OF SOME WATER-SOLUBLE PRODRUGS AND DERIVATIVES OF TAXOL WITH ANTITUMOR ACTIVITY.' cited in the application *
JOURNAL OF CONTROLLED RELEASE vol. 16, 1991, pages 121 - 136 R. DUNCAN ET AL. 'MACROMOLECULAR PRODRUGS FOR USE IN TARGETED CANCER CHEMOTHERAPY: MELPHALAN COVALENTLY COUPLED TO HMPA COPOLYMERS.' *
JOURNAL OF CONTROLLED RELEASE vol. 18, no. 2, February 1992, pages 123 - 132 V. SUBR ET AL. 'POLYMERS CONTAING ENZYMATICALLY DEGRADABLE BONDS, XII. EFFECT OF SPACER STRUCTURE ON THE RATE OF RELEASE OF DAUNOMYCIN AND ADRIAMYCIN FROM HMPA COPOLYMER DRUG CARRIERS IN VITRO AND ANTITUMOUR ACTIVITY MEASURED IN VIVO.' *
MAKROMOL. CHEM. vol. 178, 1977, pages 2169 - 2183 J. KOPECEK 'REACTIVE COPOLYMERS OF N-(2HYDROXYPROPYL)METHACRYLAMIDE WITH N-METHACRYLOYLATED DERIVATIVES OF L-LEUCINE AND L-PHENYLALANINE, 1' *

Cited By (40)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU2007254682B2 (en) * 1993-07-19 2011-06-02 Angiotech Pharmaceuticals, Inc. Anti-angiogenic compositions and methods of use
US5886026A (en) * 1993-07-19 1999-03-23 Angiotech Pharmaceuticals Inc. Anti-angiogenic compositions and methods of use
WO1995010304A1 (en) * 1993-10-08 1995-04-20 Pharmacia S.P.A Polymer-bound camptothecin derivatives
EP0693485A3 (en) * 1994-06-28 1996-05-08 Tanabe Seiyaku Co Baccatin derivatives and processes for preparing the same
US5677470A (en) * 1994-06-28 1997-10-14 Tanabe Seiyaku Co., Ltd. Baccatin derivatives and processes for preparing the same
US5608073A (en) * 1994-11-17 1997-03-04 Tanabe Seiyaku Co., Ltd. Baccatin derivatives and processes for preparing the same
EP0807115A1 (en) * 1995-01-30 1997-11-19 Enzon, Inc. High molecular weight polymer-based prodrugs
EP0807115A4 (en) * 1995-01-30 1998-10-07 Enzon Inc High molecular weight polymer-based prodrugs
US8313521B2 (en) 1995-06-07 2012-11-20 Cook Medical Technologies Llc Method of delivering an implantable medical device with a bioabsorbable coating
EP0932399A1 (en) * 1996-03-12 1999-08-04 PG-TXL Company, L.P. Water soluble paclitaxel prodrugs
EP1683520A3 (en) * 1996-03-12 2009-11-18 PG-TXL Company, L.P. Water-soluble paclitaxel produgs
EP0932399A4 (en) * 1996-03-12 2002-10-24 Pg Txl Co Lp Water soluble paclitaxel prodrugs
EP0923566A1 (en) * 1996-08-20 1999-06-23 Enzon, Inc. High molecular weight polymer-based prodrugs
EP0923566A4 (en) * 1996-08-20 2001-10-10 Enzon Inc High molecular weight polymer-based prodrugs
WO1998034968A1 (en) * 1997-02-11 1998-08-13 The Council Of The Queensland Institute Of Medical Research Polymers incorporating peptides
EP1251739A4 (en) * 1999-04-13 2004-10-06 Fannin Bioscience Inc Poly(dipeptide) as a drug carrier
EP1251739A2 (en) * 1999-04-13 2002-10-30 Fannin Bioscience, Inc. Poly(dipeptide) as a drug carrier
EP2014307A2 (en) 2001-03-13 2009-01-14 Angiotech International Ag Micellar drug delivery vehicles and uses thereof
US7875677B2 (en) 2001-04-20 2011-01-25 The University Of British Columbia Micellar drug delivery systems for hydrophobic drugs
EP2181704A2 (en) 2002-12-30 2010-05-05 Angiotech International Ag Drug delivery from rapid gelling polymer composition
US7932265B2 (en) 2004-03-31 2011-04-26 Cordis Corporation Solution formulations of sirolimus and its analogs for CAD treatment
US8557272B2 (en) 2004-03-31 2013-10-15 Cordis Corporation Device for local and/or regional delivery employing liquid formulations of therapeutic agents
US8003122B2 (en) 2004-03-31 2011-08-23 Cordis Corporation Device for local and/or regional delivery employing liquid formulations of therapeutic agents
US7846940B2 (en) 2004-03-31 2010-12-07 Cordis Corporation Solution formulations of sirolimus and its analogs for CAD treatment
US9402804B2 (en) 2004-06-02 2016-08-02 CARDINAL HEALTH SWITZERLAND 515 GmbH Injectable formulations of taxanes for cad treatment
US7989490B2 (en) 2004-06-02 2011-08-02 Cordis Corporation Injectable formulations of taxanes for cad treatment
EP2380606A1 (en) 2006-06-30 2011-10-26 Cook Incorporated Methods of manufacturing and modifying taxane coatings for implantable medical devices
EP2108390A2 (en) 2008-03-31 2009-10-14 Cordis Corporation Device for local and/or regional delivery employing liquid formulations of therapeutic agents
US8871240B2 (en) 2008-03-31 2014-10-28 Cordis Corporation Rapamycin coated expandable devices
US8409601B2 (en) 2008-03-31 2013-04-02 Cordis Corporation Rapamycin coated expandable devices
US8420110B2 (en) 2008-03-31 2013-04-16 Cordis Corporation Drug coated expandable devices
US8273404B2 (en) 2008-05-19 2012-09-25 Cordis Corporation Extraction of solvents from drug containing polymer reservoirs
EP2123312A2 (en) 2008-05-19 2009-11-25 Cordis Corporation Extraction of solvents from drug containing polymer reservoirs
US8642063B2 (en) 2008-08-22 2014-02-04 Cook Medical Technologies Llc Implantable medical device coatings with biodegradable elastomer and releasable taxane agent
US9085605B2 (en) 2010-05-27 2015-07-21 Shenzhen Salubris Pharmaceuticals Co., Ltd. Chemical synthesis and anti-tumor and anti-metastatic effects of dual functional conjugate
WO2012162010A1 (en) 2011-05-25 2012-11-29 Cordis Corporation Expandable devices coated with a paclitaxel composition
WO2012162007A1 (en) 2011-05-25 2012-11-29 Cordis Corporation Expandable devices coated with a rapamycin composition
WO2014004760A1 (en) 2012-06-28 2014-01-03 Covidien Lp Post-processing of a medical device to control morphology and mechanical properties
US9956385B2 (en) 2012-06-28 2018-05-01 The Spectranetics Corporation Post-processing of a medical device to control morphology and mechanical properties
WO2017053920A1 (en) 2015-09-25 2017-03-30 Zy Therapeutics Inc. Drug formulation based on particulates comprising polysaccharide-vitamin conjugate

Also Published As

Publication number Publication date
HUT67914A (en) 1995-05-29
AU4323393A (en) 1994-01-24
DE69330776T2 (en) 2002-07-04
HU211291A9 (en) 1995-11-28
CN1041281C (en) 1998-12-23
SG49248A1 (en) 1998-05-18
UA39926C2 (en) 2001-07-16
JP3693340B2 (en) 2005-09-07
ZA934388B (en) 1994-08-29
IL106023A (en) 1998-03-10
PL173898B1 (en) 1998-05-29
AU671247B2 (en) 1996-08-15
US5569720A (en) 1996-10-29
RU2130462C1 (en) 1999-05-20
MY109213A (en) 1996-12-31
EP0600062A1 (en) 1994-06-08
JPH06509822A (en) 1994-11-02
CN1079971A (en) 1993-12-29
KR100281606B1 (en) 2001-02-15
DE69330776D1 (en) 2001-10-25
FI940733A (en) 1994-02-16
PL302437A1 (en) 1994-07-25
US5473055A (en) 1995-12-05
CZ62094A3 (en) 1994-07-13
EP0600062B1 (en) 2001-09-19
ES2164663T3 (en) 2002-03-01
US5719265A (en) 1998-02-17
ATE205725T1 (en) 2001-10-15
CA2112482A1 (en) 1994-01-06
GB9213077D0 (en) 1992-08-05
IL106023A0 (en) 1993-10-20
AU659750B2 (en) 1995-05-25
FI940733A0 (en) 1994-02-16
HU9400800D0 (en) 1994-06-28
NZ253116A (en) 1996-05-28
TW266201B (en) 1995-12-21
MX9303598A (en) 1994-01-31
AU1628295A (en) 1995-06-22
US5362831A (en) 1994-11-08

Similar Documents

Publication Publication Date Title
US5362831A (en) Polymer-bound paclitaxel derivatives
Greenwald et al. Camptothecin-20-PEG ester transport forms: the effect of spacer groups on antitumor activity
EP0673258B1 (en) Polymer-bound camptothecin derivatives
Mellado et al. Preparation and biological activity of taxol acetates
EP0923566B1 (en) High molecular weight polymer-based prodrugs
AU705147B2 (en) High molecular weight polymer-based prodrugs
Erez et al. Enhanced cytotoxicity of a polymer–drug conjugate with triple payload of paclitaxel
CN109316605A (en) Folate receptor binding ligand-drug conjugates
CN109939242B (en) Antibody prodrug conjugate, preparation and application thereof
JP2004504358A (en) Antineoplastic polymer conjugate
KR20220038601A (en) Camptothecin drug and antibody conjugate thereof
WO1995025728A1 (en) Taxane derivatives with antitumor activity
EP2405944B1 (en) Prodrugs

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 2112482

Country of ref document: CA

AK Designated states

Kind code of ref document: A1

Designated state(s): AU BY CA CZ FI HU JP KR KZ NO NZ PL RU UA

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): AT BE CH DE DK ES FR GB GR IE IT LU MC NL PT SE

WWE Wipo information: entry into national phase

Ref document number: 1993912905

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 253116

Country of ref document: NZ

WWE Wipo information: entry into national phase

Ref document number: 940733

Country of ref document: FI

WWE Wipo information: entry into national phase

Ref document number: PV1994-620

Country of ref document: CZ

121 Ep: the epo has been informed by wipo that ep was designated in this application
WWP Wipo information: published in national office

Ref document number: 1993912905

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: PV1994-620

Country of ref document: CZ

WWG Wipo information: grant in national office

Ref document number: 1993912905

Country of ref document: EP

WWR Wipo information: refused in national office

Ref document number: PV1994-620

Country of ref document: CZ