WO1997033552A1 - Water soluble paclitaxel prodrugs - Google Patents

Water soluble paclitaxel prodrugs Download PDF

Info

Publication number
WO1997033552A1
WO1997033552A1 PCT/US1997/003687 US9703687W WO9733552A1 WO 1997033552 A1 WO1997033552 A1 WO 1997033552A1 US 9703687 W US9703687 W US 9703687W WO 9733552 A1 WO9733552 A1 WO 9733552A1
Authority
WO
WIPO (PCT)
Prior art keywords
paclitaxel
composition
acid
poly
water soluble
Prior art date
Application number
PCT/US1997/003687
Other languages
French (fr)
Inventor
Chun Li
Sidney Wallace
Dong-Fang Yu
David J. Yang
Original Assignee
Pg-Txl Company, L.P.
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 EA199800817A priority Critical patent/EA002400B1/en
Priority to KR1019980711024A priority patent/KR100561788B1/en
Priority to IL12617997A priority patent/IL126179A/en
Priority to BR9710646-1A priority patent/BR9710646A/en
Priority to HU9903952A priority patent/HU226646B1/en
Priority to JP53273497A priority patent/JP3737518B2/en
Priority to PL97328807A priority patent/PL189698B1/en
Priority to EP97917512A priority patent/EP0932399B1/en
Application filed by Pg-Txl Company, L.P. filed Critical Pg-Txl Company, L.P.
Priority to DE69735057T priority patent/DE69735057T2/en
Priority to CA002250295A priority patent/CA2250295C/en
Priority to NZ332234A priority patent/NZ332234A/en
Priority to SI9730728T priority patent/SI0932399T1/en
Priority to AU25806/97A priority patent/AU735900B2/en
Publication of WO1997033552A1 publication Critical patent/WO1997033552A1/en
Priority to UA98105316A priority patent/UA68330C2/en
Priority to NO19984210A priority patent/NO324461B1/en
Priority to NO20072562A priority patent/NO332539B1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/337Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having four-membered rings, e.g. taxol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/365Lactones
    • 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
    • A61K47/54Medicinal 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 compound
    • A61K47/547Chelates, e.g. Gd-DOTA or Zinc-amino acid chelates; Chelate-forming compounds, e.g. DOTA or ethylenediamine being covalently linked or complexed to the pharmacologically- or therapeutically-active agent
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/56Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule
    • A61K47/59Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyureas or polyurethanes
    • A61K47/60Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyureas or polyurethanes the organic macromolecular compound being a polyoxyalkylene oligomer, polymer or dendrimer, e.g. PEG, PPG, PEO or polyglycerol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/62Medicinal 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 a protein, peptide or polyamino acid
    • A61K47/64Drug-peptide, drug-protein or drug-polyamino acid conjugates, i.e. the modifying agent being a peptide, protein or polyamino acid which is covalently bonded or complexed to a therapeutically active agent
    • A61K47/645Polycationic or polyanionic oligopeptides, polypeptides or polyamino acids, e.g. polylysine, polyarginine, polyglutamic acid or peptide TAT
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K51/00Preparations containing radioactive substances for use in therapy or testing in vivo
    • A61K51/02Preparations containing radioactive substances for use in therapy or testing in vivo characterised by the carrier, i.e. characterised by the agent or material covalently linked or complexing the radioactive nucleus
    • A61K51/04Organic compounds
    • A61K51/0497Organic compounds conjugates with a carrier being an organic compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L31/00Materials for other surgical articles, e.g. stents, stent-grafts, shunts, surgical drapes, guide wires, materials for adhesion prevention, occluding devices, surgical gloves, tissue fixation devices
    • A61L31/08Materials for coatings
    • A61L31/10Macromolecular materials
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L31/00Materials for other surgical articles, e.g. stents, stent-grafts, shunts, surgical drapes, guide wires, materials for adhesion prevention, occluding devices, surgical gloves, tissue fixation devices
    • A61L31/14Materials characterised by their function or physical properties, e.g. injectable or lubricating compositions, shape-memory materials, surface modified materials
    • A61L31/16Biologically active materials, e.g. therapeutic substances
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/08Vasodilators for multiple indications
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2121/00Preparations for use in therapy
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2123/00Preparations for testing in vivo
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L2300/00Biologically active materials used in bandages, wound dressings, absorbent pads or medical devices
    • A61L2300/40Biologically active materials used in bandages, wound dressings, absorbent pads or medical devices characterised by a specific therapeutic activity or mode of action
    • A61L2300/416Anti-neoplastic or anti-proliferative or anti-restenosis or anti-angiogenic agents, e.g. paclitaxel, sirolimus
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L2300/00Biologically active materials used in bandages, wound dressings, absorbent pads or medical devices
    • A61L2300/60Biologically active materials used in bandages, wound dressings, absorbent pads or medical devices characterised by a special physical form
    • A61L2300/606Coatings

Definitions

  • the present invention relates generally to the fields of pharmaceutical compositions to be used in the treatment of cancer, autoimmune diseases and restenosis.
  • the present invention also relates to the field of pharmaceutical preparations of anticancer agents such as paclitaxel (Taxol) and doectaxel (Taxotere), in particular making paclitaxel water soluble by conjugating the drug to water soluble moieties.
  • anticancer agents such as paclitaxel (Taxol) and doectaxel (Taxotere)
  • Paclitaxel an anti-microtubule agent extracted from the needles and bark of the Pacific yew tree, Taxus brevifolia, has shown a remarkable anti-neoplastic effect in human cancer in Phase I studies and early Phase II and III trials (Horwitz et al. , 1993). This has been reported primarily in advanced ovarian and breast cancer. Significant activity has been documented in small-cell and non-small cell lung cancer, head and neck cancers, and in metastatic melanoma. However, a major difficulty in the development of paclitaxel for clinical trial use has been its insolubility in water.
  • Docetaxel is semisynthetically produced from 10-deacetyl baccatin III, a noncytotoxic precursor extracted from the needles of Taxus baccata and esterified with a chemically synthesized side chain (Cortes and Pazdur, 1995).
  • Various cancer cell lines, including breast, lung, ovarian, and colorectal cancers and melanomas have been shown to be responsive to docetaxel.
  • docetaxel has been used to achieve complete or partial responses in breast, ovarian, head and neck cancers, and malignant melanoma.
  • Paclitaxel is typically formulated as a concentrated solution containing paclitaxel 6 mg per milliliter of Cremophor EL (polyoxyethylated castor oil) and dehydrated alcohol (50% v/v) and must be further diluted before administration (Goldspiel, 1994).
  • Cremophor EL polyoxyethylated castor oil
  • the amount of Cremophor EL necessary to deliver the required doses of paclitaxel is significantly higher than that administered with any other drug that is formulated in Cremophor.
  • Cremophor EL polyoxyethylated castor oil
  • dehydrated alcohol 50% v/v
  • paclitaxel In fact, the maximum dose of paclitaxel that can be administered to mice by i.v. bolus injection is dictated by the acute lethal toxicity of the Cremophor vehicle (Eiseman et al, 1994).
  • Cremophor EL a surfactant
  • Cremophor EL a surfactant
  • DEHP di(2-ethylhexyl)phthalate
  • This preparation of paclitaxel is also shown to form paniculate matter over time and thus filtration is necessary during administration (Goldspiel, 1994). Therefore, special provisions are necessary for the preparation and administration of paclitaxel solutions to ensure safe drug delivery to patients, and these provisions inevitably lead to higher costs.
  • paclitaxel therapy Another obstacle to the widespread use of paclitaxel is the limited resources from which paclitaxel is produced, causing paclitaxel therapy to be expensive. A course of treatment may cost several thousand dollars, for example. There is the added disadvantage that not all tumors respond to paclitaxel therapy, and this may be due to the paclitaxel not getting into the tumor. There is an immediate need, therefore, for effective formulations of paclitaxel and related drugs that are water soluble with long serum half lives for treatment of tumors, autoimmune diseases such as rheumatoid arthritis, as well as for the prevention of restenosis of vessels subject to traumas such as angioplasy and stenting.
  • compositions comprising a chemotherapeutic and antiangiogenic drug, such as paclitaxel or docetaxel conjugated to a water soluble polymer such as a polyglutamic acid or a polyaspartic acid, for example, or to a water soluble metal chelator.
  • a chemotherapeutic and antiangiogenic drug such as paclitaxel or docetaxel conjugated to a water soluble polymer such as a polyglutamic acid or a polyaspartic acid, for example, or to a water soluble metal chelator.
  • These compositions are shown herein to be surprisingly effective as anti-tumor agents against exemplary tumor models, and are expected to be at least as effective as paclitaxel or docetaxel against any of the diseases or conditions for which taxanes or taxoids are known to be effective.
  • the compositions of the invention provide water soluble taxoids to overcome the drawbacks associated with the insolubility of the drugs themselves, and also provide the advantages of controlled release
  • the methods described herein could also be used to make water soluble polymer conjugates of other therapeutic agents, contrast agents and drugs, including etopside, teniposide, fludarabine, doxorubicin, daunomycin, emodin, 5-fluorouracil, FUDR, estradiol, camptothecin, retinoic acids, verapamil, epothilones and cyclosporin.
  • those agents with a free hydroxyl group would be conjugated to the polymers by similar chemical reactions as described herein for paclitaxel. Such conjugation would be well within the skill of a routine practitioner of the chemical art, and as such would fall within the scope of the claimed invention.
  • conjugated to a water soluble polymer means the covalent bonding of the drug to the polymer or chelator.
  • water soluble conjugates of the present invention may be administered in conjunction with other drugs, including other anti-tumor or anti -cancer drugs. Such combinations are known in the art.
  • the water soluble paclitaxelor docetaxel of the present invention may, in certain types of treatment, be combined with a platinum drug, an antibiotic such as doxorubicin or daunorubicin, for example, or other drugs that are used in combination with Taxol.
  • a polymer-drug conjugate that would normally remain in the vasculature may selectively leak from blood vessels into tumors, resulting in tumor accumulation of active therapeutic drug.
  • polymer-drug conjugates may act as drug depots for sustained release, producing prolonged drug exposure to tumor cells.
  • water soluble polymers may be used to stabilize drugs, as well as to solubilize otherwise insoluble compounds.
  • synthetic and natural polymers have been examined for their ability to enhance tumor- specific drug delivery (Kopecek, 1990, Maeda and Matsumura, 1989). However, only a few are currently undergoing clinical evaluation, including SMANCS in Japan and HPMA-Dox in the United Kingdom (Maeda, 1991; Kopecek and Kopeckova, 1993).
  • a taxoid is understood to mean those compounds that include paclitaxels and docetaxel, and other chemicals that have the taxane skeleton (Cortes and Pazdur, 1995), and may be isolated from natural sources such as the Yew tree, or from cell culture, or chemically synthesized molecules, and preferred is a chemical of the general chemical formula, C 47 H 51 NO 14 , including [2aR- [2a ⁇ ,4 ⁇ ,4 ⁇ ,6 ⁇ ,9 ⁇ ( ⁇ R*, ⁇ S*),l l ⁇ ,12 ⁇ ,12a ⁇ ,12b ⁇ ,]]- ⁇ -(Benzoylamino)- ⁇ - hydroxybenzenepropanoic acid 6, 12b,bis(acetyloxy)-12-(benzoyloxy)- 2a,3,4,4a,5,6,9, 10, 11 , 12, 12a, 12b-dodecahydro-4, 11 -dihydroxy-4a,8, 13 , 13- tetramethyl-5-oxo-7,l l-
  • paclitaxel and docetaxel are each more effective than the other against certain types of tumors, and that in the practice of the present invention, those tumors that are more susceptible to a particular taxoid would be treated with that water soluble taxoid conjugate.
  • the composition may further comprise a chelated metal ion.
  • the chelated metal ion of the present invention may be an ionic form of any one of aluminum, boron, calcium, chromium, cobalt, copper, dysprosium, erbium, europium, gadolinium, gallium, germanium, holmium, indium, iridium, iron, magnesium, manganese, nickel, platinum, rhenium, rubidium, ruthenium, samarium, sodium, technetium, thallium, tin, yttrium or zinc.
  • the chelated metal ion will be a radionuclide, i.e., a radioactive isotope of one of the listed ⁇ * 7 Aft t i l Q m_ metals.
  • Preferred radionuclides include, but are not limited to Ga, Ga, In, Tc,
  • Preferred water soluble chelators to be used in the practice of the present invention include, but are not limited to, diethylenetriaminepentaacetic acid (DTP A), ethylenediaminetetraacetic acid (EDTA), 1,4,7,10-tetraazacyclododecane- N,N',N",N'"-tetraacetate (DOTA), tetraazacyclotetradecane-N,N',N"N , "-tetraacetic acid (TETA), hydroxyethylidene diphosphonate (HEDP), dimercaptosuccinic acid (DMSA), diethylenetriaminetetramethylenephosphonic acid (DTTP) and l-(p- aminobenzyl)-DTPA, 1,6-diamino hexane-N,N,N',N' -tetraacetic acid, DPDP, and ethylenebis (oxyethylenenitrilo)-tetraacetic acid, with DTPA being the most
  • the paclitaxel or docetaxel may be conjugated to a water soluble polymer, and preferably the polymer is conjugated to the 2' or the 7- hydroxyl or both of the paclitaxel or docetaxel.
  • a degradable linkage in this case, an ester, is used to ensure that the active drug is released from the polymeric carrier.
  • Preferred polymers include, but are not limited to polyethylene glycol, poly(l-glutamic acid), poly(d-glutamic acid), poly(dl-glutamic acid), poly(l-aspartic acid), poly(d-aspartic acid), poly(dl-aspartic acid), polyethylene glycol, copolymers of the above listed polyamino acids with polyethylene glycol, polycaprolactone, polyglycolic acid and polylactic acid, as well as polyacrylic acid, poly(2-hydroxyethyl 1-glutamine), carboxymethyl dextran, hyaluronic acid, human serum albumin and alginic acid, with polyethylene glycol, polyaspartic acids and polyglutamic acids being particularly preferred.
  • the polyglutamic acids or polyaspartic acids of the present invention preferably have a molecular weight of about 5,000 to about 100,000 with about 20,000 to about 80,000, or even about 30,000 to about 60,000 being more preferred. It is understood that the compositions of the present invention may be dispersed in a pharmaceutically acceptable carrier solution as described below. Such a solution would be sterile or aseptic and may include water, buffers, isotonic agents or other ingredients known to those of skill in the art that would cause no allergic or other harmful reaction when administered to an animal or human subject.
  • the present invention may also be described as a pharmaceutical composition
  • a pharmaceutical composition comprising a chemotherapeutic or anti-cancer drug such as paclitaxel or docetaxel conjugated to a high molecular weight water soluble polymer or to a chelator.
  • the pharmaceutical composition may include polyethylene glycol, polyglutamatic acids polyaspartatic acids or a chelator, preferably DTPA.
  • a radionuclide may be used as an anti-tumor agent, or drug, and that the present pharmaceutical composition may include a therapeutic amount of a chelated radioactive isotope.
  • the present invention may be described as a method of determining the uptake of a chemotherapeutic drug such as paclitaxel or docetaxel by tumor tissue.
  • This method may comprise obtaining a conjugate of the drug and a metal chelator with a chelated metal ion, contacting tumor tissue with the composition and detecting the presence of the chelated metal ion in the tumor tissue.
  • the presence of the chelated metal ion in the tumor tissue is indicative of uptake by the tumor tissue.
  • the chelated metal ion may be a radionuclide and the detection may be scintigraphic.
  • the tumor tissue may also be contained in an animal or a human subject and the composition would then be administered to the subject.
  • the present invention may also be described in certain embodiments as a method of treating cancer in a subject.
  • This method includes obtaining a composition comprising a chemotherapeutic drug such as paclitaxel or docetaxel conjugated to a water soluble polymer or chelator and dispersed in a pharmaceutically acceptable solution and administering the solution to the subject in an amount effective to treat the tumor.
  • chemotherapeutic drug such as paclitaxel or docetaxel conjugated to a water soluble polymer or chelator and dispersed in a pharmaceutically acceptable solution
  • Preferred compositions comprise paclitaxel or docetaxel conjugated to a polyglutamic acids or polyaspartic acids and more preferably to poly (1-glutamic acid) or poly 1-aspartic acid).
  • compositions of the invention are understood to be effective against any type of cancer for which the unconjugated taxoid is shown to be effective and would include, but not be limited to breast cancer, ovarian cancer, malignant melanoma, lung cancer, gastric cancer, colon cancer, head and neck cancer or leukemia.
  • the method of treating a tumor may include some prediction of the paclitaxel or docetaxel uptake in the tumor prior to administering a therapeutic amount of the drug or prodrug.
  • This method may include any of the imaging techniques discussed above in which a paclitaxel-chelator-chelated metal is administered to a subject and detected in a tumor. This step provides a cost effective way of determining that a particular tumor would not be expected to respond to DTPA-paclitaxel therapy in those cases where the drug does not get into the tumor. It is contemplated that if an imaging technique can be used to predict the response to paclitaxel and to identify patients that are not likely to respond, great expense and crucial time may be saved for the patient. The assumption is that if there is no reasonable amount of chemotherapeutic agent deposited in the tumor, the probability of tumor response to that agent is relatively small.
  • the present invention may be described as a method of obtaining a body image of a subject.
  • the body image is obtained by administering an effective amount of a radioactive metal ion chelated to a paclitaxel-chelator conjugate to a subject and measuring the scintigraphic signals of the radioactive metal to obtain an image.
  • the present invention may also be described in certain broad aspects as a method of decreasing at least one symptom of a systemic autoimmune disease comprising administering to a subject having a systemic autoimmune disease an effective amount of a composition comprising paclitaxel or docetaxel conjugated to poly-1-glutamic acid or poly-1-aspartic acid.
  • a composition comprising paclitaxel or docetaxel conjugated to poly-1-glutamic acid or poly-1-aspartic acid.
  • the effectiveness of the water soluble taxoids of the present invention will not be diminished by the conjugation to a water soluble moiety, and that the water soluble prodrug may act as a controlled release formulation that releases the active drug over a period of time. Therefore, the compositions of the present invention are expected to be as effective as Taxol against rheumatoid arthritis, for example, but will offer the advantage a controlled release. It is also understood that the taxoid compositions of the present invention may be used in combination with other drugs, such as an angiogenesis inhibitor (AGM-1470) (Oliver et al. , 1994) or methotrexate.
  • AGM-1470 an angiogenesis inhibitor
  • methotrexate methotrexate
  • water soluble paclitaxels and docetaxels of the present invention will find a variety of applications beyond direct parenteral administration (WO 9625176).
  • water soluble paclitaxel will be useful as a coating for implanted medical devices, such as tubings, shunts, catheters, artificial implants, pins, electrical implants such as pacemakers, and especially for arterial or venous stents, including balloon-expandable stents.
  • water soluble paclitaxel may be bound to an implantable medical device, or alternatively, the water soluble paclitaxel may be passively adsorbed to the surface of the implantable device.
  • stents may be coated with polymer- drug conjugates by dipping the ste in polymer-drug solution or spraying the stent with such a solution.
  • Suitable materials for the implantable device should be biocompatible and nontoxic and may be chosen from the metals such as nickel- titanium alloys, steel, or biocompatible polymers, hydrogels, polyurethanes, polyethylenes, ethylenevinyl acetate copolymers, etc.
  • the water soluble paclitaxel is coated onto a stent for insertion into an artery or vein following balloon angioplasty.
  • the invention may be described therefore, in certain broad aspects as a method of inhibiting arterial restenosis or arterial occlusion following vascular trauma comprising administering to a subject in need thereof, a composition comprising paclitaxel or docetaxel conjugated to poly-1-glutamic acid or poly-1-aspartic acid.
  • the subject may be a coronary bypass, vascular surgery, organ transplant or coronary or arterial angioplasty patient, for example, and the composition may be administered directly, intravenously, or even coated on a stent and the stent is implanted at the sight of vascular trauma.
  • An embodiment of the invention is, therefore, an implantable medical device, wherein the device is coated with a composition comprising paclitaxel or docetaxel conjugated to polyglutamic acids or polyaspartic acids in an amount effective to inhibit smooth muscle cell proliferation.
  • a preferred device is a stent coated with the compositions of the present invention as described herein, and in certain preferred embodiments, the stent is adapted to be used after balloon angioplasty and the coating is effective to inhibit restenosis.
  • the invention may be described as a composition comprising polyglutamic acids conjugated to the 2' or 7 hydroxyl or both of paclitaxel, or even a composition comprising polyaspartic acid conjugated to the 2' or 7 hydroxyl or both of paclitaxel.
  • a polyglutamic acid or “polyglutamic acids” include poly (1-glutamic acid), poly (d-glutamic acid) and poly (dl-glutamic acid)
  • a polyaspartic acid” or “polyaspartic acids” include poly (1-aspartic acid), poly (d-aspartic acid) and poly (dl-aspartic acid).
  • FIG. 1A Chemical structure of paclitaxel, PEG-paclitaxel and DTPA-paclitaxel.
  • FIG. IB Chemical structure and reaction scheme for production of PG-paclitaxel.
  • FIG. 2 Effect of paclitaxel, PEG-paclitaxel and DTPA-paclitaxel on proliferation of B16 melanoma cells.
  • FIG. 3 Antitumor effect of DTPA-paclitaxel on MCa-4 mammary tumors.
  • FIG. 4 Median time (days) to reach tumor diameter of 12 mm after treatment with paclitaxel, DTPA-paclitaxel and PEG-paclitaxel.
  • FIG. 5 Gamma-scintigraphs of mice bearing MCa-4 tumors following intravenous injection of In-DTPA-paclitaxel and 'in-DTPA. Arrow indicates the tumor.
  • FIG. 6 Hydrolytic degradation of PG-paclitaxel as determined in PBS at pH 7.4 at 37 C.
  • — ⁇ -- represents percent paclitaxel remaining attached to soluble PG
  • represents percent paclitaxel released
  • — O ⁇ represents percent metabolite- 1 produced.
  • FIG. 7A The antitumor effect of PG-paclitaxel on rats bearing murine breast tumor (13762F).
  • -D- represents the response to a single i.v. dose of PG (0.3 g/kg);
  • - ⁇ - represents response to paclitaxel (40 mg/kg),
  • -O- represents response to PG-paclitaxel (60 mg equiv. paclitaxel/kg).
  • FIG. 7B The antitumor effect of PG-paclitaxel and paclitaxel on mice bearing OCa-1 tumors.
  • -D- represents the response to a single i.v. dose of PG (0.8 g/kg);
  • - ⁇ - represents response to paclitaxel (80 mg/kg),
  • -•- represents response to PG-paclitaxel (80 mg equiv. paclitaxel/kg),
  • -O- represents response to PG-paclitaxel (160 mg equiv. paclitaxel/kg).
  • FIG. 7C The antitumor effect of PG-paclitaxel on mice bearing MCa-4 mammary carcinoma tumors.
  • -D- represents the response to a single i.v.
  • - ⁇ - represents the response to a single i.v. dose of PG (0.6 g/kg); - ⁇ - represents response to PG-paclitaxel (40 mg/kg), -0- represents response to PG-paclitaxel (60 mg equiv. paclitaxel/kg), -O- represents response to PG-paclitaxel (120 mg/kg).
  • FIG. 7D The antitumor effect of PG-paclitaxel against soft-tissue sarcoma tumor (FSa-II) in mice.
  • -D- represents the response to a single i.v. dose of saline
  • -0- represents the response to a single i.v. dose of PG (0.8 g/kg)
  • -O- represents response to paclitaxel (80 mg/kg)
  • - ⁇ - represents response to PG-paclitaxel (160 mg equiv. paclitaxel/kg).
  • FIG. 7E The antitumor effect of PG-paclitaxel against syngeneic hepatocarcinoma tumor (HCa-I) in mice.
  • -D- represents the response to a single i.v. dose of saline
  • - ⁇ - represents the response to a single i.v. dose of PG (0.8 g/kg)
  • -O- represents response to PG-paclitaxel (80 mg/kg)
  • - ⁇ - represents response to PG-paclitaxel (160 mg equiv. paclitaxel/kg).
  • FIG. 8 Release profile of paclitaxel from PEG-paclitaxel in phosphate buffer (pH 7.4). Paclitaxel, -X-; PEG-paclitaxel, -O-.
  • FIG. 9 Antitumor effect of PEG-paclitaxel on MCa-4 mammary tumors.
  • -G- represents the response a single i.v. injection with a saline solution of PEG (60 mg/ml)
  • - ⁇ - represents the response to the Cremophor/alcohol vehicle
  • -O- represents a single dose of 40 mg/kg body weight of paclitaxel
  • -•- represents PEG-paclitaxel at 40 mg equiv. paclitaxel/kg body weight.
  • the present invention arises from the discovery of novel, water soluble formulations of paclitaxel and docetaxel, and the surprising efficacy of these formulations against tumor cells in vivo.
  • Poly (1-glutamic acid) conjugated paclitaxel (PG-paclitaxel) administered to mice bearing ovarian carcinoma (OCa-I) caused significant tumor growth delay as compared to the same dose of paclitaxel without PG.
  • Mice treated with paclitaxel alone or with a combination of free paclitaxel and PG showed delayed tumor growth initially, but tumors regrew to levels comparable to an untreated control group after ten days.
  • MTD maximal tolerated dose
  • PG- paclitaxel is one of the most stable water-soluble paclitaxel derivatives known (Deutsch, et al, 1989; Mathew et al, 1992; Zhao and Scientific, 1991).
  • DTPA-paclitaxel is also shown herein to be as effective as paclitaxel in an in vitro antitumor potency assay using a B16 melanoma cell line.
  • DTPA-paclitaxel did not show any significant difference in antitumor effect as compared to paclitaxel against an MCa-4 mammary tumor at a dose of 40 mg/kg body weight in a single injection.
  • Indium labeled DTPA-paclitaxel was shown to accumulate in the MCa-4 tumor as demonstrated by gamma-scintigraphy, demonstrating that the chelator conjugated anti-tumor drugs of the present invention are useful and effective for tumor imaging.
  • novel compounds and methods of the present invention provide significant advances over prior methods and compositions, as the water-soluble paclitaxels are projected to improve the efficacy of paclitaxel-based anti-cancer therapy, by providing water soluble and controlled release paclitaxel derived compositions.
  • Such compositions eliminate the need for solvents that are associated with side effects seen with prior paclitaxel compositions.
  • radiolabeled paclitaxel which is shown to retain anti-tumor activity, will also be useful in the imaging of tumors.
  • the present invention allows one to determine whether a paclitaxel will be taken up by a particular tumor by scintigraphy, single photon emission computer tomography (SPECT) or positron emission tomography (PET). This determination may then be used to decide the efficacy of anti-cancer treatment. This information may be helpful in guiding the practitioner in the selection of patients to undergo paclitaxel therapy.
  • SPECT single photon emission computer tomography
  • PET positron emission tomography
  • the paclitaxel may be rendered water-soluble in two ways: by conjugating paclitaxel to water-soluble polymers which serve as drug carriers, and by derivatizing the antitumor drug with water soluble chelating agents.
  • the latter approach also provides an opportunity for labeling with radionuclides (e.g., ' In, Y, Ho, Ga, 99m Tc) for nuclear imaging and/or for radiotherapy studies.
  • radionuclides e.g., ' In, Y, Ho, Ga, 99m Tc
  • the structures of paclitaxel, polyethylene glycol-paclitaxel (PEG-paclitaxel), polyglutamic acid- paclitaxel conjugate (PG-paclitaxel) and diethylenetriaminepentaacetic acid-paclitaxel (DTPA-paclitaxel) are shown in FIG. 1.
  • DTPA-paclitaxel or other paclitaxel-chelating agent conjugates such as EDTA-paclitaxel, DTTP-paclitaxel, or DOTA-paclitaxel, for example, may be prepared in the form of water-soluble salts (sodium salt, potassium salt, tetrabutylammonium salt, calcium salt, ferric salt, etc.). These salts will be useful as therapeutic agents for tumor treatment. Secondly, DTPA-paclitaxel or other paclitaxel-chelating agents will be useful as diagnostic agents which, when labeled with radionuclides such as In or m Tc, may be used as radiotracers to detect certain tumors in combination with nuclear imaging techniques.
  • radionuclides such as In or m Tc
  • taxane derivatives may be adapted for use in the compositions and methods of the present invention and that all such compositions and methods would be encompassed by the appended claims.
  • a stent coated with the polymer- paclitaxel conjugates may be used to prevent restenosis, the closure of arteries following balloon angioplasty.
  • Recent results in clinical trials using balloon- expandable stents in coronary angioplasty have shown a significant benefit in patency and the reduction of restenosis compared to standard balloon angioplasty (Serruys et al, 1994).
  • neointima formation is associated with increased cell proliferation.
  • SMC smooth muscle cell
  • anti-cancer drugs may be useful to prevent neointimal SMC accumulation.
  • Stents coated with polymer-linked anti-proliferative agents that are capable of releasing these agents over a prolonged period of time with sufficient concentration will thus prevent ingrowth of hyperplastic intima and media into the lumen thereby reducing restenosis.
  • the formulations of the present invention are also contemplated to be useful in the treatment of autoimmune and or inflammatory diseases such as rheumatoid arthritis
  • Paclitaxel binding to tubulin shifts the equilibrium to stable microtubender polymers and makes this drug a strong inhibitor of eukaryotic cell replication by blocking cells in the late G2 mitotic stage.
  • Several mechanisms may be involved in arthritis suppression by paclitaxel.
  • paclitaxel's phase specific cytotoxic effects may affect rapidly proliferating inflammatory cells, and furthermore paclitaxel inhibits, cell mitosis, migration, chemotaxis, intracellular transport and neutrophil H 2 O 2 production.
  • paclitaxel may have antiangiogenic activity by blocking coordinated endothelial cell migration (Oliver et al, 1994). Therefore, the polymer conjugated prodrugs of the present invention are contemplated to be as useful as free paclitaxel in the treatment of rheumatoid arthritis.
  • the polymer conjugated formulation disclosed herein would also offer the advantages of delayed or sustained release of the drug and greater solubility. It is also an aspect of the treatment of arthritis that the formulations may be injected or implanted directly into the affected joint areas.
  • the pharmaceutical preparations of paclitaxel or docetaxel suitable for injectable use include sterile aqueous solutions or dispersions and sterile powders for the preparation of sterile injectable solutions or dispersions.
  • the form must be sterile and must be fluid for injection. It must be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms, such as bacteria and fungi.
  • the carrier may be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyethylene glycol, and the like), suitable mixtures thereof, and vegetable oils.
  • microorganisms can be brought about by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, thimerosal, and the like.
  • antibacterial and antifungal agents for example, parabens, chlorobutanol, phenol, sorbic acid, thimerosal, and the like.
  • isotonic agents for example, sugars or sodium chloride.
  • Sterile injectable solutions are prepared by incorporating the active compounds in the required amount in the appropriate solvent with various of the other ingredients enumerated above, as required, followed by filtered sterilization.
  • dispersions are prepared by incorporating the various sterilized active ingredients into a sterile vehicle which contains the basic dispersion medium and the required other ingredients from those enumerated above.
  • the preferred methods of preparation are vacuum-drying and freeze-drying techniques which yield a powder of the active ingredient plus any additional desired ingredient from a previously sterile- filtered solution thereof.
  • pharmaceutically acceptable carrier includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents and isotonic agents and the like.
  • the use of such media and agents for pharmaceutically active substances is well known in the art. Except insofar as any conventional media or agent is incompatible with the active ingredient, its use in the therapeutic compositions is contemplated. Supplementary active ingredients can also be incorporated into the compositions.
  • phrases “pharmaceutically acceptable” also refers to molecular entities and compositions that do not produce an allergic or similar untoward reaction when administered to an animal or a human.
  • aqueous solution for parenteral administration in an aqueous solution, for example, the solution should be suitably buffered if necessary and the liquid diluent first rendered isotonic with sufficient saline or glucose.
  • aqueous solutions are especially suitable for intravenous and intraperitoneal administration.
  • sterile aqueous media which can be employed will be known to those of skill in the art in light of the present disclosure.
  • DTPA-paclitaxel To a solution of paclitaxel (100 mg, 0.117 mmol) in dry DMF (2.2 ml) was added diethylenetriaminepentaacetic acid anhydride (DTPA A) (210 mg, 0.585 mmol) at 0°C. The reaction mixture was stirred at 4°C overnight. The suspension was filtered (0.2 ⁇ m Millipore filter) to remove unreacted DTPA anhydride. The filtrate was poured into distilled water, stirred at 4°C for 20 min, and the precipitate collected. The crude product was purified by preparative TLC over C 18 silica gel plates and developed in acetonitrile/water (1 :1).
  • DTPA A diethylenetriaminepentaacetic acid anhydride
  • Paclitaxel had an R f value of 0.34.
  • the band above the paclitaxel with an R f value of 0.65 to 0.75 was removed by scraping and eluted with an acetonitrile/water (1 :1) mixture, and the solvent was removed to give 15 mg of DTPA-paclitaxel as product (yield 10.4%): mp: > 226 °C dec.
  • the UV spectrum (sodium salt in water) showed maximal absorption at 228 nm which is also characteristic for paclitaxel.
  • the sodium salt of DTPA-paclitaxel was also obtained by adding a solution of DTPA-paclitaxel in ethanol into an equivalent amount of 0.05 M NaHCO 3 , followed by lyophilizing to yield a water-soluble solid powder (solubility>20 mg equivalent paclitaxel/ml). Hydrolytic stability of DTPA-paclitaxel:
  • DTPA-paclitaxel The hydrolytic stability of DTPA-paclitaxel was studied under accelerated conditions. Briefly, 1 mg of DTPA-paclitaxel was dissolved in 1 ml 0.5 M NaHCO 3 aqueous solution (pH 9.3) and analyzed by HPLC.
  • HPLC system consisted of a Waters 150 x 3.9 (i.d.) mm Nova-Pak column filled with C18 4 ⁇ m silica gel, a Perkin-Elmer isocratic LC pump, a PE Nelson 900 series interface, a Spectra-Physics UV/Vis detector and a data station.
  • the retention times of DTPA-paclitaxel and paclitaxel were 1.38 and 8.83 min, respectively. Peak areas were quantitated and compared with standard curves to determine the DTPA-paclitaxel and paclitaxel concentrations.
  • the estimated half-life of DTPA-paclitaxel in 0.5 M NaHCO 3 solution is about 16 days at room temperature.
  • Cells were seeded in 24-well plates at a concentration of 2.5 x 10 cells/ml and grown in a 50:50 Dulbecco's modified minimal essential medium (DEM) and F12 medium containing 10% bovine calf serum at 37°C for 24 hrs in a 97% humidified atmosphere of 5.5% CO 2 . The medium was then replaced with fresh medium containing paclitaxel or DTPA-paclitaxel in concentration ranging from 5 x 10 "9 M to 75 x 10 " M. After 40 hrs, the cells were released by trypsinization and counted in a Coulter counter.
  • DEM Dulbecco's modified minimal essential medium
  • F12 medium containing 10% bovine calf serum at 37°C for 24 hrs in a 97% humidified atmosphere of 5.5% CO 2 .
  • the medium was then replaced with fresh medium containing paclitaxel or DTPA-paclitaxel in concentration ranging from 5 x 10 "9 M to 75 x 10 " M. After 40
  • the final concentrations of DMSO (used to dissolve paclitaxel) and 0.05 M sodium bicarbonate solution (used to dissolve DTPA-paclitaxel) in the cell medium were less than 0.01%. This amount of solvent did not have any effect on cell growth as determined by control studies.
  • DTPA-paclitaxel The effects of DTPA-paclitaxel on the growth of B16 melanoma cells are presented in FIG. 2. After a 40-h incubation with various concentrations, DTPA- paclitaxel and paclitaxel were compared as to cytotoxicity. The IC 50 for paclitaxel and DTPA-paclitaxel are 15 nM and 7.5 nM, respectively. Antitumor effect on mammary carcinoma (MCa-4) tumor model:
  • mice Female C3Hf/Kam mice were inoculated with mammary carcinoma (MCa-4) in the muscles of the right thigh (5 x 10 cells/mouse). When the tumors had grown to 8 mm (approx. 2 wks), a single dose of paclitaxel or DTPA-paclitaxel was given at 10. 20 and 40 mg equivalent paclitaxel/kg body weight. In control studies, saline and absolute alcohol/Cremophor 50/50 diluted with saline (1 :4) were used. Tumor growth was determined daily, by measuring three orthogonal tumor diameters. When the tumor size reached 12 mm in diameter, the tumor growth delay was calculated. The mice were sacrificed when tumors were approximately 15 mm.
  • MCa-4 mammary carcinoma
  • the tumor growth curve is shown in FIG. 3.
  • both paclitaxel and DTPA-paclitaxel showed antitumor effect at a dose of 40 mg/kg.
  • the data were also analyzed to determine the mean number of days for the tumor to reach 12 mm in diameter.
  • Statistical analysis showed that DTPA-paclitaxel delayed tumor growth significantly compared to the saline treated control at a dose of 40 mg/kg (p ⁇ 0.01).
  • the mean time for the tumor to reach 12 mm in diameter was 12.1 days for DTPA-paclitaxel compared to 9.4 days for paclitaxel (FIG. 4).
  • HPLC HPLC was used to analyze the reaction mixture and purity of In-DTPA-paclitaxel.
  • the system consisted of a LDC binary pump, a 100 x 8.0 mm (i.d.) Waters column filled with ODS 5 ⁇ m silica gel. The column was eluted at a flow rate of 1 ml/min with a gradient mixture of water and methanol (gradient from 0% to 85% methanol over 15 min). The gradient system was monitored with a Nal crystal detector and a Spectra-Physics UV Vis detector.
  • mice Female C3Hf7Kam mice were inoculated with mammary carcinoma (MCa-4) in the muscles of the right thigh (5 x 10 5 cells). When the tumors had grown to 12 mm in diameter, the mice were divided into two groups. In group I, the mice were anesthetized by intraperitoneal injection of sodium pentobarbital, followed by In- DTPA-paclitaxel (100-200 mCi) via tail vein. A ⁇ -camera equipped with a medium energy collimator was positioned over the mice (3 per group). A series of 5 min acquisitions were collected at 5, 30, 60, 120, 240 min and 24 hrs after injection. In group II, the same procedures were followed except that the mice were injected with mammary carcinoma (MCa-4) in the muscles of the right thigh (5 x 10 5 cells). When the tumors had grown to 12 mm in diameter, the mice were divided into two groups. In group I, the mice were anesthetized by intraperitoneal injection of sodium pentobarbital, followed
  • FIG. 5 shows gamma-scintigraphs of animals injected with "in-DTPA and '" In-DTPA-paclitaxel.
  • ⁇ l In-DTPA was characterized by rapid clearance from the plasma, rapid and high excretion in the urine with minimal retention in the kidney and negligible retention in the tumor, the liver, the intestine and other organs or body parts.
  • In-DTPA-paclitaxel exhibited a pharmacological profile resembling that of paclitaxel (Eiseman et al, 1994). Radioactivity in the brain was negligible. Liver and kidney had the greatest tissue:plasma ratios.
  • the present example demonstrates the conjugation of paclitaxel to a water- soluble polymer, poly (1-glutamic acid) (PG).
  • PG poly (1-glutamic acid
  • the potential of water-soluble polymers used as drug carriers is well established (Kopecek, 1990; Maeda and Matsumura, 1989).
  • the drug-polymer conjugate also acts as a slow-release depot for controlled drug release.
  • PG was selected as a carrier for paclitaxel because it can be readily degraded by lysosomal enzymes, is stable in plasma and contains sufficient functional groups for drug attachment.
  • antitumor drugs including Adriamycin (Van Heeswijk et al, 1985; Hoes et al, 1985), cyclophosphamide (Hirano et al, 1979), and Ara-C
  • PG sodium salt (MW 34 K, Sigma, 0.35 g) was dissolved in water. The pH of the aqueous solution was adjusted to 2 using 0.2 M HCI. The precipitate was collected, dialyzed against distilled water, and lyophilized to yield 0.29 g PG.
  • the sodium salt of PG-paclitaxel conjugate was obtained by dissolving the product in 0.5 M NaHCO 3 .
  • the aqueous solution of PG-paclitaxel was dialyzed against distilled water (MWCO 1 ,000) to remove low molecular weight contaminants and excess NaHCO 3 salt. Lyophilization of the dialysate yielded 88.6 mg of white powder.
  • the paclitaxel content in this polymeric conjugate was determined by UV (described below) as 21 % (w/w). Yield (conversion to polymer bound paclitaxel, UV): 93%.
  • PG-paclitaxel with higher paclitaxel content (up to 35%) can be synthesized by this method by simply increasing the ratio of paclitaxel to PG used.
  • UV spectra were obtained on a Beckman DU-640 spectrophotometer (Fullerton, CA).
  • PG-paclitaxel has characteristic paclitaxel absorption with ⁇ shifts from 228 to 230 nm.
  • the concentration of paclitaxel in PG-paclitaxel was estimated based on standard curve generated with known concentrations of paclitaxel in methanol at absorption of 228 nm, assuming that the polymer conjugate in water at 230 nm and the free drug in methanol at 228 nm have the same molar extinction and both follow Lambert Beer's law.
  • the relative molecular weight of PG-paclitaxel was characterized by gel permeation chromatography (GPC).
  • GPC gel permeation chromatography
  • the GPC system consisted of two LDC model III pumps coupled with LDC gradient master, a PL gel GPC column, and a Waters 990 photodiode array detector.
  • the elutant (DMF) was run at 1.0 ml/min with UV detection set at 270 nm. Conjugation of paclitaxel to PG resulted in an increase in the molecular weight of PG-paclitaxel, as indicated by the shift of retention time from 6.4 min of PG to 5.0 min of PG-paclitaxel conjugate as analyzed by GPC.
  • the calculated molecular weight of PG-paclitaxel containing 15-25% paclitaxel is in the range of 45-55 kDa.
  • the crude product contained a small molecular weight contaminant (retention time 8.0 to 10.0 min, and 11.3 min), which can be effectively removed by converting PG-paclitaxel to its sodium salt, followed by dialysis.
  • the HPLC system consisted of a reversed-phase silica column (Nova-Pac, Waters, CA), a mobile phase of methanol-water (2:1, v/v) delivered at a flow rate of 1.0 ml min, and a photodiode detector.
  • concentration of PG bound paclitaxel, free paclitaxel and other degradation products in each sample was calculated by comparing the peak areas with a separately obtained standard curve prepared from paclitaxel, assuming that the molar extinction coefficient of each peak at 228 nm is the same as that of paclitaxel.
  • the half-life of the conjugate estimated to be 132, 40 and 4 days at pH 6.0, 7.4 and 9.6 respectively, were determined by a linear least-square regression analysis.
  • HPLC analysis revealed that incubation of PG-paclitaxel in PBS solutions produced paclitaxel and several other species including one that is more hydrophobic than paclitaxel (metabolite- 1).
  • the amount of metabolite- 1, which was most probably 7-epipaclitaxel, recovered in PBS at pH 7.4 surpassed that of paclitaxel after 100 hours of incubation (FIG. 6).
  • tubulin polymerization assay Aliquots obtained from PBS solution at pH 7.4 were subjected to analysis by a tubulin polymerization assay.
  • the tubulin assembly reaction was performed at 32°C in PEM buffer (pH 6.9) at a tubulin (bovine brain, Cytoskeleton Inc., Boulder, CO) concentration of 1 mg/ml (10 ⁇ M) in the presence of testing samples (1.0 ⁇ M equiv. Paclitaxel) and 1.0 mM GTP.
  • Tubulin polymerization was followed by measuring the absorbance of the solution at 340 nm over time. After 15 min, calcium chloride (125 mM) was added to measure CaCl 2 -induced depolymerization of microtubules.
  • MTT tetrazolium salt
  • PG-paclitaxel inhibited tumor cell proliferation to an extent similar to that of free paclitaxel.
  • the resultant IC 50 values were 0.59 ⁇ M for paclitaxel and 0.82 ⁇ M for PG-paclitaxel (measured in paclitaxel equivalent units).
  • IC 50 6.79 ⁇ M
  • the IC 50 of PG alone was greater than 100 ⁇ M.
  • C3H/Kam mice were bred and maintained in a pathogen-free facility in the Department of Experimental Radiation Oncology. Solitary tumors were produced in the muscle of the right thigh of female C3H/Kam mice (25-30g) by injecting 5 x 10 murine ovarian carcinoma cells (OCa-I), mammary carcinoma (MCa-4), hepatocarcinoma (HCa-I) or fibrous sarcoma (FSa-II).
  • OCa-I murine ovarian carcinoma cells
  • MCa-4 mammary carcinoma
  • HCa-I hepatocarcinoma
  • FSa-II fibrous sarcoma
  • female Fischer 344 rats 125-150 g were injected with 1.0 x 10 5 viable 13762F tumor cells in 0.1 ml PBS.
  • PG-paclitaxel solution was prepared by dissolving the conjugate in saline (8 mg equiv. paclitaxel/ml). The injected volume at 60 mg/kg was 0.975 ml per rat.
  • Paclitaxel Cremophor solution was prepared by dissolving paclitaxel in a 1 :1 mixture of ethyl alcohol and Cremophor (30 mg/ml). This stock solution was further diluted with saline (1 :4) before injection. The final concentration of paclitaxel in the solution was 6 mg/ml. The injected volume at 60 mg/kg was
  • PG solution was prepared by dissolving the polymer in saline (22 mg/ml).
  • the injected dose was 0.3 g/kg (1.8 ml per rat), which was equivalent to paclitaxel dose of 60 mg/kg.
  • Cremophor vehicle was prepared by diluting a mixture of ethyl alcohol and cremophor (1 :1) with saline (1 :4).
  • PG-paclitaxel 40-120 mg equiv. paclitaxel/kg in saline or paclitaxel in Cremophor vehicle i.v. in a single injection.
  • Control animals were treated with saline (0.6 ml), Cremophor vehicle (0.5 ml), PG solution in saline, or PG g/kg) plus paclitaxel (80 mg/kg).
  • b Tumor growth was determined by daily measurement of three orthogonal diameters with calipers and the volume was calculated as (a x b x c)/2. Shown in brackets are the number of mice used in each group. The time in days to grow from 500 mm to 2000 mm are presented mean ⁇ standard deviation.
  • Absolute growth delay defined as the time in days for tumors treated with various drugs to grow from 500 to 2000 mm 3 minus the time in days for tumors treated with saline control to grow from 500 to 2000 mm 3 , d
  • the time in days to grow from 500 to 2000 mm were compared for treatment groups and saline group using Student's /-Test. P- values are two-sided and were taken to be significant when less than to equal 0.05.
  • PEG-paclitaxel polyethylene glycol-paclitaxel
  • the synthesis was accomplished in two steps. First 2'-succinyl-paclitaxel was prepared according to a reported procedure (Deutsch et al, 1989). Paclitaxel (200 mg, 0.23 mmol) and succinic anhydride (288 mg, 2.22 mmol) were allowed to react in anhydrous pyridine (6 ml) at room temperature for 3 hrs. The pyridine was then evaporated, and the residue was treated with water, stirred for 20 min, and filtered. The precipitate was dissolved in acetone, water was slowly added, and the fine crystals were collected to yield 180 mg 2'-succinyl-paclitaxel.
  • PEG-paclitaxel was synthesized by an N-ethoxycarbonyl-2-ethoxy-l,2-dihydroquinoline (EEDQ) mediated coupling reaction.
  • EEDQ N-ethoxycarbonyl-2-ethoxy-l,2-dihydroquinoline
  • PEG-Paclitaxel Hydrolytic stability of PEG-Paclitaxel
  • PEG-Paclitaxel was dissolved in phosphate buffer (0.01M) at various pHs at a concentration of 0.4 mM and the solutions were allowed to incubate at 37°C with gentle shaking. At selected time intervals, aliquots (200 ⁇ l) were removed and lyophilized. The resulting dry powders were redissolved in methylene chloride for gel permeation chromatography (GPC analysis).
  • the GPC system consisted of a Perkin- Elmer PL gel mixed bed column, a Perkin-Elmer isocratic LC pump, a PE Nelson 900 series interface, a Spectra-Physics UV/Vis detector and a data station.
  • the elutant (methylene chloride) was run at 1.0 ml/min with the UV detector set at 228 nm.
  • the retention times of PEG-paclitaxel and paclitaxel were 6.1 and 8.2 min, respectively. Peak areas were quantified and the percentage of PEG-paclitaxel remaining and the percentage of paclitaxel released were calculated.
  • the half life of PEG-paclitaxel determined by linear least-squares at pH 7.4 was 54 min.
  • the half-life at pH 9.0 was 7.6 min. Release profiles of paclitaxel from PEG-paclitaxel at pH 7.4 is shown in FIG. 8.
  • the cells were released by trypsinization and counted in a Coulter counter.
  • the final concentrations of DMSO (used to dissolve paclitaxel) and 0.05 M sodium bicarbonate solution (used to dissolve PEG-paclitaxel) in the cell medium were less than 0.01%. This amount of solvent did not have any effect on cell growth as determined by control studies. Furthermore, PEG in the concentration range used to generate an equivalent paclitaxel concentration from 5 x 10 "9 M to 75 x 10 "9 M also did not effect cell proliferation.
  • MCa-4 cells (5 x 10 s cells) were injected into the right thigh muscle of female C3Hf/Kam mice.
  • a single dose of paclitaxel or PEG- paclitaxel was given at 10, 20 and at 40 mg equivalent paclitaxel/kg body weight.
  • Paclitaxel was initially dissolved in absolute ethanol with an equal volume of Cremophor.
  • This stock solution was further diluted (1 :4 by volume) with a sterile physiological solution within 15 minutes of injection.
  • PEG-paclitaxel was dissolved in saline (6 mg equiv. paclitaxel/ml) and filtered through a sterile filter (Millipore, 4.5 ⁇ m).
  • Saline, paclitaxel vehicle, absolute alcohol: Cremophor (1 : 1) diluted with saline (1 :4) and PEG solution in saline (600 mg/kg body weight) were used in control experiments.
  • Tumor growth was determined daily, by measuring three orthogonal tumor diameters. When the tumor size reached 12 mm in diameter, the tumor growth delay was calculated.
  • the tumor growth curve is shown in FIG 9.
  • both PEG- paclitaxel and paclitaxel effectively delayed tumor growth.
  • Paclitaxel was more effective than PEG-paclitaxel, although the difference was not statistically significant.
  • Paclitaxel treated tumors required 9.4 days to reach 12 mm in diameter whereas PEG- paclitaxel-treated tumors required 8.5 days.
  • these values were significant (p > 0.05) as compared to their corresponding controls, which were 6.7 days for the paclitaxel vehicle and 6.5 days for the saline solution of PEG (FIG. 4).

Abstract

Disclosed are water soluble compositions of paclitaxel and docetaxel formed by conjugating the paclitaxel or docetaxel to a water soluble chelator, polyethylene glycol or polymer such as poly (1-glutamic acid) or poly (1-aspartic acid). Also disclosed are methods of using the compositions for treatment of tumors, autoimmune disorders such as rheumatoid arthritis and for prediction of paclitaxel uptake by tumors and radiolabeled DTPA-paclitaxel tumor imaging. Other embodiments include the coating of implantable stents for prevention of restenosis.

Description

DESCRIPTION
WATER SOLUBLE PACLITAXEL PRODRUGS
FIELD OF THE INVENTION
The present invention relates generally to the fields of pharmaceutical compositions to be used in the treatment of cancer, autoimmune diseases and restenosis. The present invention also relates to the field of pharmaceutical preparations of anticancer agents such as paclitaxel (Taxol) and doectaxel (Taxotere), in particular making paclitaxel water soluble by conjugating the drug to water soluble moieties.
BACKGROUND OF THE INVENTION Paclitaxel, an anti-microtubule agent extracted from the needles and bark of the Pacific yew tree, Taxus brevifolia, has shown a remarkable anti-neoplastic effect in human cancer in Phase I studies and early Phase II and III trials (Horwitz et al. , 1993). This has been reported primarily in advanced ovarian and breast cancer. Significant activity has been documented in small-cell and non-small cell lung cancer, head and neck cancers, and in metastatic melanoma. However, a major difficulty in the development of paclitaxel for clinical trial use has been its insolubility in water.
Docetaxel is semisynthetically produced from 10-deacetyl baccatin III, a noncytotoxic precursor extracted from the needles of Taxus baccata and esterified with a chemically synthesized side chain (Cortes and Pazdur, 1995). Various cancer cell lines, including breast, lung, ovarian, and colorectal cancers and melanomas have been shown to be responsive to docetaxel. In clinical trials, docetaxel has been used to achieve complete or partial responses in breast, ovarian, head and neck cancers, and malignant melanoma.
Paclitaxel is typically formulated as a concentrated solution containing paclitaxel 6 mg per milliliter of Cremophor EL (polyoxyethylated castor oil) and dehydrated alcohol (50% v/v) and must be further diluted before administration (Goldspiel, 1994). The amount of Cremophor EL necessary to deliver the required doses of paclitaxel is significantly higher than that administered with any other drug that is formulated in Cremophor. Several toxic effects have been attributed to Cremophor, including vasodilation, dyspnea, and hypotension. This vehicle has also been shown to cause serious hypersensitivity in laboratory animals and humans (Weiss et al , 1990). In fact, the maximum dose of paclitaxel that can be administered to mice by i.v. bolus injection is dictated by the acute lethal toxicity of the Cremophor vehicle (Eiseman et al, 1994). In addition, Cremophor EL, a surfactant, is known to leach phthalate plasticizers such as di(2-ethylhexyl)phthalate (DEHP) from the polyvinylchloride bags and intravenous administration tubing. DEHP is known to cause hepatotoxicity in animals and is carcinogenic in rodents. This preparation of paclitaxel is also shown to form paniculate matter over time and thus filtration is necessary during administration (Goldspiel, 1994). Therefore, special provisions are necessary for the preparation and administration of paclitaxel solutions to ensure safe drug delivery to patients, and these provisions inevitably lead to higher costs.
Prior attempts to obtain water soluble paclitaxel have included the preparation of prodrugs of paclitaxel by placing solubilizing moieties such as succinate and amino acids at the 2'-hydroxyl group or at the 7-hydroxyl position (Deutsch et al, 1989; Mathew et al, 1992). However, these prodrugs have not proven chemically stable enough for development. For example, Deutsch et al. (1989) report a 2'-succinate derivative of paclitaxel, but water solubility of the sodium salt is only about 0.1% and the triethanolamine and N-methylglucamine salts were soluble at only about 1%. In addition, amino acid esters were reponed to be unstable. Similar results were reported by Mathew et al. (1992). Greenwald et al. reported the synthesis of highly water- soluble 2' and 7-polyethylene glycol esters of taxol (Greenwald et al, 1994). however, no data concerning the in vivo antitumor activity of these compounds were reported (Greenwald et al. 1995). Others attempts to solve these problems have involved microencapsulation of paclitaxel in both liposomes and nanospheres (Bartoni and Boitard, 1990). The liposome formulation was reported to be as effective as free paclitaxel, however only liposome formulations containing less than 2% paclitaxel were physically stable (Sharma and Straubinger, 1994). Unfortunately, the nanosphere formulation proved to be toxic. There is still a need therefore for a water soluble paclitaxel formulation that can deliver effective amounts of paclitaxel and docetaxel without the disadvantages caused by the insolubility of the drug.
Another obstacle to the widespread use of paclitaxel is the limited resources from which paclitaxel is produced, causing paclitaxel therapy to be expensive. A course of treatment may cost several thousand dollars, for example. There is the added disadvantage that not all tumors respond to paclitaxel therapy, and this may be due to the paclitaxel not getting into the tumor. There is an immediate need, therefore, for effective formulations of paclitaxel and related drugs that are water soluble with long serum half lives for treatment of tumors, autoimmune diseases such as rheumatoid arthritis, as well as for the prevention of restenosis of vessels subject to traumas such as angioplasy and stenting.
SUMMARY OF THE INVENTION
The present invention seeks to overcome these and other drawbacks inherent in the prior art by providing compositions comprising a chemotherapeutic and antiangiogenic drug, such as paclitaxel or docetaxel conjugated to a water soluble polymer such as a polyglutamic acid or a polyaspartic acid, for example, or to a water soluble metal chelator. These compositions are shown herein to be surprisingly effective as anti-tumor agents against exemplary tumor models, and are expected to be at least as effective as paclitaxel or docetaxel against any of the diseases or conditions for which taxanes or taxoids are known to be effective. The compositions of the invention provide water soluble taxoids to overcome the drawbacks associated with the insolubility of the drugs themselves, and also provide the advantages of controlled release so that tumors are shown herein to be eradicated in animal models after a single intravenous administration.
The methods described herein could also be used to make water soluble polymer conjugates of other therapeutic agents, contrast agents and drugs, including etopside, teniposide, fludarabine, doxorubicin, daunomycin, emodin, 5-fluorouracil, FUDR, estradiol, camptothecin, retinoic acids, verapamil, epothilones and cyclosporin. In particular, those agents with a free hydroxyl group would be conjugated to the polymers by similar chemical reactions as described herein for paclitaxel. Such conjugation would be well within the skill of a routine practitioner of the chemical art, and as such would fall within the scope of the claimed invention. Those agents would include, but would not be limited to etopside, teniposide, camptothecin and the epothilones. As used herein, conjugated to a water soluble polymer means the covalent bonding of the drug to the polymer or chelator.
It is also understood that the water soluble conjugates of the present invention may be administered in conjunction with other drugs, including other anti-tumor or anti -cancer drugs. Such combinations are known in the art. The water soluble paclitaxelor docetaxel of the present invention may, in certain types of treatment, be combined with a platinum drug, an antibiotic such as doxorubicin or daunorubicin, for example, or other drugs that are used in combination with Taxol.
Conjugation of chemotherapeutic drugs to polymers is an attractive approach to reduce systemic toxicity and improve the therapeutic index. Polymers with molecular mass larger than 30 kDa do not readily diffuse through normal capillaries and glomerular endothelium, thus sparing normal tissue from irrelevant drug-mediated toxicity (Maeda and Matsumura, 1989; Reynolds. 1995). On the other hand, it is well established that malignant tumors often have disordered capillary endothelium and greater permeability than normal tissue vasculature (Maeda and Matsumura, 1989; Fidler et al, 1987). Thus, a polymer-drug conjugate that would normally remain in the vasculature may selectively leak from blood vessels into tumors, resulting in tumor accumulation of active therapeutic drug. Additionally, polymer-drug conjugates may act as drug depots for sustained release, producing prolonged drug exposure to tumor cells. Finally, water soluble polymers may be used to stabilize drugs, as well as to solubilize otherwise insoluble compounds. At present, a variety of synthetic and natural polymers have been examined for their ability to enhance tumor- specific drug delivery (Kopecek, 1990, Maeda and Matsumura, 1989). However, only a few are currently undergoing clinical evaluation, including SMANCS in Japan and HPMA-Dox in the United Kingdom (Maeda, 1991; Kopecek and Kopeckova, 1993).
In the present disclosure, a taxoid is understood to mean those compounds that include paclitaxels and docetaxel, and other chemicals that have the taxane skeleton (Cortes and Pazdur, 1995), and may be isolated from natural sources such as the Yew tree, or from cell culture, or chemically synthesized molecules, and preferred is a chemical of the general chemical formula, C47H51 NO14, including [2aR- [2aα,4β,4αβ,6β,9α(αR*,βS*),l lα,12α,12aα,12bα,]]-β-(Benzoylamino)-α- hydroxybenzenepropanoic acid 6, 12b,bis(acetyloxy)-12-(benzoyloxy)- 2a,3,4,4a,5,6,9, 10, 11 , 12, 12a, 12b-dodecahydro-4, 11 -dihydroxy-4a,8, 13 , 13- tetramethyl-5-oxo-7,l l-methano-lH-cyclodeca[3,4]benz-[l,2-b]oxet-9-yl ester. It is understood that paclitaxel and docetaxel are each more effective than the other against certain types of tumors, and that in the practice of the present invention, those tumors that are more susceptible to a particular taxoid would be treated with that water soluble taxoid conjugate.
In those embodiments in which the paclitaxel is conjugated to a water soluble metal chelator, the composition may further comprise a chelated metal ion. The chelated metal ion of the present invention may be an ionic form of any one of aluminum, boron, calcium, chromium, cobalt, copper, dysprosium, erbium, europium, gadolinium, gallium, germanium, holmium, indium, iridium, iron, magnesium, manganese, nickel, platinum, rhenium, rubidium, ruthenium, samarium, sodium, technetium, thallium, tin, yttrium or zinc. In certain preferred embodiments, the chelated metal ion will be a radionuclide, i.e., a radioactive isotope of one of the listed Λ*7 Aft t i l Q m_ metals. Preferred radionuclides include, but are not limited to Ga, Ga, In, Tc,
90Y, l l4mIn and 193mPt.
Preferred water soluble chelators to be used in the practice of the present invention include, but are not limited to, diethylenetriaminepentaacetic acid (DTP A), ethylenediaminetetraacetic acid (EDTA), 1,4,7,10-tetraazacyclododecane- N,N',N",N'"-tetraacetate (DOTA), tetraazacyclotetradecane-N,N',N"N,"-tetraacetic acid (TETA), hydroxyethylidene diphosphonate (HEDP), dimercaptosuccinic acid (DMSA), diethylenetriaminetetramethylenephosphonic acid (DTTP) and l-(p- aminobenzyl)-DTPA, 1,6-diamino hexane-N,N,N',N' -tetraacetic acid, DPDP, and ethylenebis (oxyethylenenitrilo)-tetraacetic acid, with DTPA being the most preferred. A preferred embodiment of the present invention may also be a composition comprising "in-DTPA-paclitaxel.
In certain embodiments of the present invention, the paclitaxel or docetaxel may be conjugated to a water soluble polymer, and preferably the polymer is conjugated to the 2' or the 7- hydroxyl or both of the paclitaxel or docetaxel. Thus when functional groups are used for drug conjugation, as above with the C2'-hydroxyl of paclitaxel, a degradable linkage, in this case, an ester, is used to ensure that the active drug is released from the polymeric carrier. Preferred polymers include, but are not limited to polyethylene glycol, poly(l-glutamic acid), poly(d-glutamic acid), poly(dl-glutamic acid), poly(l-aspartic acid), poly(d-aspartic acid), poly(dl-aspartic acid), polyethylene glycol, copolymers of the above listed polyamino acids with polyethylene glycol, polycaprolactone, polyglycolic acid and polylactic acid, as well as polyacrylic acid, poly(2-hydroxyethyl 1-glutamine), carboxymethyl dextran, hyaluronic acid, human serum albumin and alginic acid, with polyethylene glycol, polyaspartic acids and polyglutamic acids being particularly preferred. The polyglutamic acids or polyaspartic acids of the present invention preferably have a molecular weight of about 5,000 to about 100,000 with about 20,000 to about 80,000, or even about 30,000 to about 60,000 being more preferred. It is understood that the compositions of the present invention may be dispersed in a pharmaceutically acceptable carrier solution as described below. Such a solution would be sterile or aseptic and may include water, buffers, isotonic agents or other ingredients known to those of skill in the art that would cause no allergic or other harmful reaction when administered to an animal or human subject. Therefore, the present invention may also be described as a pharmaceutical composition comprising a chemotherapeutic or anti-cancer drug such as paclitaxel or docetaxel conjugated to a high molecular weight water soluble polymer or to a chelator. The pharmaceutical composition may include polyethylene glycol, polyglutamatic acids polyaspartatic acids or a chelator, preferably DTPA. It is also understood that a radionuclide may be used as an anti-tumor agent, or drug, and that the present pharmaceutical composition may include a therapeutic amount of a chelated radioactive isotope.
In certain embodiments, the present invention may be described as a method of determining the uptake of a chemotherapeutic drug such as paclitaxel or docetaxel by tumor tissue. This method may comprise obtaining a conjugate of the drug and a metal chelator with a chelated metal ion, contacting tumor tissue with the composition and detecting the presence of the chelated metal ion in the tumor tissue. The presence of the chelated metal ion in the tumor tissue is indicative of uptake by the tumor tissue. The chelated metal ion may be a radionuclide and the detection may be scintigraphic. The tumor tissue may also be contained in an animal or a human subject and the composition would then be administered to the subject.
The present invention may also be described in certain embodiments as a method of treating cancer in a subject. This method includes obtaining a composition comprising a chemotherapeutic drug such as paclitaxel or docetaxel conjugated to a water soluble polymer or chelator and dispersed in a pharmaceutically acceptable solution and administering the solution to the subject in an amount effective to treat the tumor. Preferred compositions comprise paclitaxel or docetaxel conjugated to a polyglutamic acids or polyaspartic acids and more preferably to poly (1-glutamic acid) or poly 1-aspartic acid). The compositions of the invention are understood to be effective against any type of cancer for which the unconjugated taxoid is shown to be effective and would include, but not be limited to breast cancer, ovarian cancer, malignant melanoma, lung cancer, gastric cancer, colon cancer, head and neck cancer or leukemia.
The method of treating a tumor may include some prediction of the paclitaxel or docetaxel uptake in the tumor prior to administering a therapeutic amount of the drug or prodrug. This method may include any of the imaging techniques discussed above in which a paclitaxel-chelator-chelated metal is administered to a subject and detected in a tumor. This step provides a cost effective way of determining that a particular tumor would not be expected to respond to DTPA-paclitaxel therapy in those cases where the drug does not get into the tumor. It is contemplated that if an imaging technique can be used to predict the response to paclitaxel and to identify patients that are not likely to respond, great expense and crucial time may be saved for the patient. The assumption is that if there is no reasonable amount of chemotherapeutic agent deposited in the tumor, the probability of tumor response to that agent is relatively small.
In certain embodiments the present invention may be described as a method of obtaining a body image of a subject. The body image is obtained by administering an effective amount of a radioactive metal ion chelated to a paclitaxel-chelator conjugate to a subject and measuring the scintigraphic signals of the radioactive metal to obtain an image.
The present invention may also be described in certain broad aspects as a method of decreasing at least one symptom of a systemic autoimmune disease comprising administering to a subject having a systemic autoimmune disease an effective amount of a composition comprising paclitaxel or docetaxel conjugated to poly-1-glutamic acid or poly-1-aspartic acid. Of particular interest in the context of the present disclosure is the treatment of rheumatoid arthritis, which is known to respond in some cases to taxol when administered in the standard Cremophor formulation (US Patent 5,583,153). As in the treatment of tumors, it is contemplated that the effectiveness of the water soluble taxoids of the present invention will not be diminished by the conjugation to a water soluble moiety, and that the water soluble prodrug may act as a controlled release formulation that releases the active drug over a period of time. Therefore, the compositions of the present invention are expected to be as effective as Taxol against rheumatoid arthritis, for example, but will offer the advantage a controlled release. It is also understood that the taxoid compositions of the present invention may be used in combination with other drugs, such as an angiogenesis inhibitor (AGM-1470) (Oliver et al. , 1994) or methotrexate.
The finding that paclitaxel also inhibits restenosis after balloon angioplasty indicates that the water soluble paclitaxels and docetaxels of the present invention will find a variety of applications beyond direct parenteral administration (WO 9625176). For example, it is contemplated that water soluble paclitaxel will be useful as a coating for implanted medical devices, such as tubings, shunts, catheters, artificial implants, pins, electrical implants such as pacemakers, and especially for arterial or venous stents, including balloon-expandable stents. In these embodiments it is contemplated that water soluble paclitaxel may be bound to an implantable medical device, or alternatively, the water soluble paclitaxel may be passively adsorbed to the surface of the implantable device. For example, stents may be coated with polymer- drug conjugates by dipping the ste in polymer-drug solution or spraying the stent with such a solution. Suitable materials for the implantable device should be biocompatible and nontoxic and may be chosen from the metals such as nickel- titanium alloys, steel, or biocompatible polymers, hydrogels, polyurethanes, polyethylenes, ethylenevinyl acetate copolymers, etc. In a preferred embodiment the water soluble paclitaxel, especially a PG-paclitaxel conjugate, is coated onto a stent for insertion into an artery or vein following balloon angioplasty. The invention may be described therefore, in certain broad aspects as a method of inhibiting arterial restenosis or arterial occlusion following vascular trauma comprising administering to a subject in need thereof, a composition comprising paclitaxel or docetaxel conjugated to poly-1-glutamic acid or poly-1-aspartic acid. In the practice of the method, the subject may be a coronary bypass, vascular surgery, organ transplant or coronary or arterial angioplasty patient, for example, and the composition may be administered directly, intravenously, or even coated on a stent and the stent is implanted at the sight of vascular trauma.
An embodiment of the invention is, therefore, an implantable medical device, wherein the device is coated with a composition comprising paclitaxel or docetaxel conjugated to polyglutamic acids or polyaspartic acids in an amount effective to inhibit smooth muscle cell proliferation. A preferred device is a stent coated with the compositions of the present invention as described herein, and in certain preferred embodiments, the stent is adapted to be used after balloon angioplasty and the coating is effective to inhibit restenosis.
In certain preferred embodiments, the invention may be described as a composition comprising polyglutamic acids conjugated to the 2' or 7 hydroxyl or both of paclitaxel, or even a composition comprising polyaspartic acid conjugated to the 2' or 7 hydroxyl or both of paclitaxel. As used herein, the terms "a polyglutamic acid" or "polyglutamic acids" include poly (1-glutamic acid), poly (d-glutamic acid) and poly (dl-glutamic acid) and the terms "a polyaspartic acid" or "polyaspartic acids" include poly (1-aspartic acid), poly (d-aspartic acid) and poly (dl-aspartic acid).
Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. Although any methods and materials similar or equivalent to those described herein can be used in the practice or testing of the present invention, the preferred methods and materials are now described.
BRIEF DESCRIPTION OF THE DRAWINGS FIG. 1A. Chemical structure of paclitaxel, PEG-paclitaxel and DTPA-paclitaxel. FIG. IB. Chemical structure and reaction scheme for production of PG-paclitaxel.
FIG. 2. Effect of paclitaxel, PEG-paclitaxel and DTPA-paclitaxel on proliferation of B16 melanoma cells.
FIG. 3. Antitumor effect of DTPA-paclitaxel on MCa-4 mammary tumors.
FIG. 4. Median time (days) to reach tumor diameter of 12 mm after treatment with paclitaxel, DTPA-paclitaxel and PEG-paclitaxel.
FIG. 5. Gamma-scintigraphs of mice bearing MCa-4 tumors following intravenous injection of In-DTPA-paclitaxel and 'in-DTPA. Arrow indicates the tumor.
FIG. 6. Hydrolytic degradation of PG-paclitaxel as determined in PBS at pH 7.4 at 37 C. — □-- represents percent paclitaxel remaining attached to soluble PG, ~Δ~ represents percent paclitaxel released, — O ~ represents percent metabolite- 1 produced.
FIG. 7A. The antitumor effect of PG-paclitaxel on rats bearing murine breast tumor (13762F). -D- represents the response to a single i.v. dose of PG (0.3 g/kg); -Δ- represents response to paclitaxel (40 mg/kg), -O- represents response to PG-paclitaxel (60 mg equiv. paclitaxel/kg).
FIG. 7B. The antitumor effect of PG-paclitaxel and paclitaxel on mice bearing OCa-1 tumors. -D- represents the response to a single i.v. dose of PG (0.8 g/kg); -Δ- represents response to paclitaxel (80 mg/kg), -•- represents response to PG-paclitaxel (80 mg equiv. paclitaxel/kg), -O- represents response to PG-paclitaxel (160 mg equiv. paclitaxel/kg). FIG. 7C. The antitumor effect of PG-paclitaxel on mice bearing MCa-4 mammary carcinoma tumors. -D- represents the response to a single i.v. dose of saline, -Δ- represents the response to a single i.v. dose of PG (0.6 g/kg); -♦- represents response to PG-paclitaxel (40 mg/kg), -0- represents response to PG-paclitaxel (60 mg equiv. paclitaxel/kg), -O- represents response to PG-paclitaxel (120 mg/kg).
FIG. 7D. The antitumor effect of PG-paclitaxel against soft-tissue sarcoma tumor (FSa-II) in mice. -D- represents the response to a single i.v. dose of saline, -0- represents the response to a single i.v. dose of PG (0.8 g/kg); -O- represents response to paclitaxel (80 mg/kg), -Δ- represents response to PG-paclitaxel (160 mg equiv. paclitaxel/kg).
FIG. 7E. The antitumor effect of PG-paclitaxel against syngeneic hepatocarcinoma tumor (HCa-I) in mice. -D- represents the response to a single i.v. dose of saline, -Δ- represents the response to a single i.v. dose of PG (0.8 g/kg); -O- represents response to PG-paclitaxel (80 mg/kg), -Δ- represents response to PG-paclitaxel (160 mg equiv. paclitaxel/kg).
FIG. 8. Release profile of paclitaxel from PEG-paclitaxel in phosphate buffer (pH 7.4). Paclitaxel, -X-; PEG-paclitaxel, -O-.
FIG. 9. Antitumor effect of PEG-paclitaxel on MCa-4 mammary tumors. -G- represents the response a single i.v. injection with a saline solution of PEG (60 mg/ml), -■- represents the response to the Cremophor/alcohol vehicle, -O-represents a single dose of 40 mg/kg body weight of paclitaxel, -•- represents PEG-paclitaxel at 40 mg equiv. paclitaxel/kg body weight.
DETAILED DESCRIPTION OF THE INVENTION
The present invention arises from the discovery of novel, water soluble formulations of paclitaxel and docetaxel, and the surprising efficacy of these formulations against tumor cells in vivo. Poly (1-glutamic acid) conjugated paclitaxel (PG-paclitaxel) administered to mice bearing ovarian carcinoma (OCa-I) caused significant tumor growth delay as compared to the same dose of paclitaxel without PG. Mice treated with paclitaxel alone or with a combination of free paclitaxel and PG showed delayed tumor growth initially, but tumors regrew to levels comparable to an untreated control group after ten days. Moreover, at the maximal tolerated dose (MTD) of the PG-paclitaxel conjugate, (160 mg equiv. paclitaxel/kg), the growth of tumors was completely suppressed, the tumors shrank, and mice observed for two months following treatment remained tumor free (MTD: defined as the maximal dose that produced 15% or less body weight loss within two weeks after a single i.v. injection). In a parallel study, the antitumor activity of PG-paclitaxel in rats with rat mammary adenocarcinoma (13762F) was examined. Again, complete tumor eradication at 40-60 mg equiv. paclitaxel/kg of PG-paclitaxel was observed. These surprising results demonstrate that the polymer-drug conjugate, PG-paclitaxel, successfully eradicates well established solid tumors in both mice and rats after a single intravenous injection. Moreover, with a half-life of 40 days at pH 7.4, PG- paclitaxel is one of the most stable water-soluble paclitaxel derivatives known (Deutsch, et al, 1989; Mathew et al, 1992; Zhao and Kingston, 1991).
DTPA-paclitaxel is also shown herein to be as effective as paclitaxel in an in vitro antitumor potency assay using a B16 melanoma cell line. DTPA-paclitaxel did not show any significant difference in antitumor effect as compared to paclitaxel against an MCa-4 mammary tumor at a dose of 40 mg/kg body weight in a single injection. Furthermore, Indium labeled DTPA-paclitaxel was shown to accumulate in the MCa-4 tumor as demonstrated by gamma-scintigraphy, demonstrating that the chelator conjugated anti-tumor drugs of the present invention are useful and effective for tumor imaging.
The novel compounds and methods of the present invention provide significant advances over prior methods and compositions, as the water-soluble paclitaxels are projected to improve the efficacy of paclitaxel-based anti-cancer therapy, by providing water soluble and controlled release paclitaxel derived compositions. Such compositions eliminate the need for solvents that are associated with side effects seen with prior paclitaxel compositions. In addition, radiolabeled paclitaxel, which is shown to retain anti-tumor activity, will also be useful in the imaging of tumors. Further, the present invention allows one to determine whether a paclitaxel will be taken up by a particular tumor by scintigraphy, single photon emission computer tomography (SPECT) or positron emission tomography (PET). This determination may then be used to decide the efficacy of anti-cancer treatment. This information may be helpful in guiding the practitioner in the selection of patients to undergo paclitaxel therapy.
The paclitaxel may be rendered water-soluble in two ways: by conjugating paclitaxel to water-soluble polymers which serve as drug carriers, and by derivatizing the antitumor drug with water soluble chelating agents. The latter approach also provides an opportunity for labeling with radionuclides (e.g., ' In, Y, Ho, Ga, 99mTc) for nuclear imaging and/or for radiotherapy studies. The structures of paclitaxel, polyethylene glycol-paclitaxel (PEG-paclitaxel), polyglutamic acid- paclitaxel conjugate (PG-paclitaxel) and diethylenetriaminepentaacetic acid-paclitaxel (DTPA-paclitaxel) are shown in FIG. 1.
In certain embodiments of the present invention, DTPA-paclitaxel or other paclitaxel-chelating agent conjugates, such as EDTA-paclitaxel, DTTP-paclitaxel, or DOTA-paclitaxel, for example, may be prepared in the form of water-soluble salts (sodium salt, potassium salt, tetrabutylammonium salt, calcium salt, ferric salt, etc.). These salts will be useful as therapeutic agents for tumor treatment. Secondly, DTPA-paclitaxel or other paclitaxel-chelating agents will be useful as diagnostic agents which, when labeled with radionuclides such as In or mTc, may be used as radiotracers to detect certain tumors in combination with nuclear imaging techniques. It is understood that in addition to paclitaxel (taxol) and docetaxel (taxotere), other taxane derivatives may be adapted for use in the compositions and methods of the present invention and that all such compositions and methods would be encompassed by the appended claims.
Toxicity studies, pharmacokinetics and tissue distribution of DTPA-paclitaxel have shown that in mice the LD50 (50% lethal dose) of DPTA-paclitaxel observed with a single dose intravenous (iv) injection is about 110 mg/kg body weight. Direct comparison with paclitaxel is difficult to make because of the dose-volume restraints imposed by limited solubility of paclitaxel and vehicle toxicity associated with iv administration. However, in light of the present disclosure, one skilled in the art of chemotherapy would determine the effective and maximal tolerated dosages in a clinical study for use in human subjects.
In certain embodiments of the invention, a stent coated with the polymer- paclitaxel conjugates may be used to prevent restenosis, the closure of arteries following balloon angioplasty. Recent results in clinical trials using balloon- expandable stents in coronary angioplasty have shown a significant benefit in patency and the reduction of restenosis compared to standard balloon angioplasty (Serruys et al, 1994). According to the response-to-injury hypothesis, neointima formation is associated with increased cell proliferation. Currently, popular opinion holds that the critical process leading to vascular lesions in both spontaneous and accelerated atherosclerosis is smooth muscle cell (SMC) proliferation (Phillips-Hughes and Kandarpa, 1996). Since SMC phenotypic proliferation after arterial injury mimics that of neoplastic cells, it is possible that anti-cancer drugs may be useful to prevent neointimal SMC accumulation. Stents coated with polymer-linked anti-proliferative agents that are capable of releasing these agents over a prolonged period of time with sufficient concentration will thus prevent ingrowth of hyperplastic intima and media into the lumen thereby reducing restenosis.
Because paclitaxel has been shown to suppress collagen induced arthritis in a mouse model (Oliver et al, 1994), the formulations of the present invention are also contemplated to be useful in the treatment of autoimmune and or inflammatory diseases such as rheumatoid arthritis Paclitaxel binding to tubulin shifts the equilibrium to stable microtubiile polymers and makes this drug a strong inhibitor of eukaryotic cell replication by blocking cells in the late G2 mitotic stage. Several mechanisms may be involved in arthritis suppression by paclitaxel. For example, paclitaxel's phase specific cytotoxic effects may affect rapidly proliferating inflammatory cells, and furthermore paclitaxel inhibits, cell mitosis, migration, chemotaxis, intracellular transport and neutrophil H2O2 production. In addition, paclitaxel may have antiangiogenic activity by blocking coordinated endothelial cell migration (Oliver et al, 1994). Therefore, the polymer conjugated prodrugs of the present invention are contemplated to be as useful as free paclitaxel in the treatment of rheumatoid arthritis. The polymer conjugated formulation disclosed herein would also offer the advantages of delayed or sustained release of the drug and greater solubility. It is also an aspect of the treatment of arthritis that the formulations may be injected or implanted directly into the affected joint areas.
The pharmaceutical preparations of paclitaxel or docetaxel suitable for injectable use include sterile aqueous solutions or dispersions and sterile powders for the preparation of sterile injectable solutions or dispersions. In all cases the form must be sterile and must be fluid for injection. It must be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms, such as bacteria and fungi. The carrier may be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyethylene glycol, and the like), suitable mixtures thereof, and vegetable oils. The prevention of the action of microorganisms can be brought about by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, thimerosal, and the like. In many cases, it will be preferable to include isotonic agents, for example, sugars or sodium chloride.
Sterile injectable solutions are prepared by incorporating the active compounds in the required amount in the appropriate solvent with various of the other ingredients enumerated above, as required, followed by filtered sterilization. Generally, dispersions are prepared by incorporating the various sterilized active ingredients into a sterile vehicle which contains the basic dispersion medium and the required other ingredients from those enumerated above. In the case of sterile powders for the preparation of sterile injectable solutions, the preferred methods of preparation are vacuum-drying and freeze-drying techniques which yield a powder of the active ingredient plus any additional desired ingredient from a previously sterile- filtered solution thereof.
As used herein, "pharmaceutically acceptable carrier" includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents and isotonic agents and the like. The use of such media and agents for pharmaceutically active substances is well known in the art. Except insofar as any conventional media or agent is incompatible with the active ingredient, its use in the therapeutic compositions is contemplated. Supplementary active ingredients can also be incorporated into the compositions.
The phrase "pharmaceutically acceptable" also refers to molecular entities and compositions that do not produce an allergic or similar untoward reaction when administered to an animal or a human.
For parenteral administration in an aqueous solution, for example, the solution should be suitably buffered if necessary and the liquid diluent first rendered isotonic with sufficient saline or glucose. These particular aqueous solutions are especially suitable for intravenous and intraperitoneal administration. In this connection, sterile aqueous media which can be employed will be known to those of skill in the art in light of the present disclosure.
The following examples are included to demonstrate preferred embodiments of the invention. It should be appreciated by those of skill in the art that the techniques disclosed in the examples which follow represent techniques discovered by the inventor to function well in the practice of the invention, and thus can be considered to constitute preferred modes for its practice. However, those of skill in the art should, in light of the present disclosure, appreciate that many changes can be made in the specific embodiments which are disclosed and still obtain a like or similar result without departing from the spirit and scope of the invention.
Example 1 DTPA-Paclitaxel
Synthesis of DTPA-paclitaxel: To a solution of paclitaxel (100 mg, 0.117 mmol) in dry DMF (2.2 ml) was added diethylenetriaminepentaacetic acid anhydride (DTPA A) (210 mg, 0.585 mmol) at 0°C. The reaction mixture was stirred at 4°C overnight. The suspension was filtered (0.2 μm Millipore filter) to remove unreacted DTPA anhydride. The filtrate was poured into distilled water, stirred at 4°C for 20 min, and the precipitate collected. The crude product was purified by preparative TLC over C18 silica gel plates and developed in acetonitrile/water (1 :1). Paclitaxel had an Rf value of 0.34. The band above the paclitaxel with an Rf value of 0.65 to 0.75 was removed by scraping and eluted with an acetonitrile/water (1 :1) mixture, and the solvent was removed to give 15 mg of DTPA-paclitaxel as product (yield 10.4%): mp: > 226 °C dec. The UV spectrum (sodium salt in water) showed maximal absorption at 228 nm which is also characteristic for paclitaxel. Mass spectrum: (FAB) m/e 1229 (M+H)+, 1251 (M+Na), 1267 (M+K). In the Η NMR spectrum (DMSO-d6) the resonance of NCH2CH2N and CH COOH of DTPA appeared as a complex series of signals at δ 2.71 - 2.96 ppm, and as a multiplet at δ 3.42 ppm, respectively. The resonance of C7- H at 4.10 ppm in paclitaxel shifted to 5.51 ppm, suggesting esterification at the 7- position. The rest of the spectrum was consistent with the structure of paclitaxel.
The sodium salt of DTPA-paclitaxel was also obtained by adding a solution of DTPA-paclitaxel in ethanol into an equivalent amount of 0.05 M NaHCO3, followed by lyophilizing to yield a water-soluble solid powder (solubility>20 mg equivalent paclitaxel/ml). Hydrolytic stability of DTPA-paclitaxel:
The hydrolytic stability of DTPA-paclitaxel was studied under accelerated conditions. Briefly, 1 mg of DTPA-paclitaxel was dissolved in 1 ml 0.5 M NaHCO3 aqueous solution (pH 9.3) and analyzed by HPLC. The HPLC system consisted of a Waters 150 x 3.9 (i.d.) mm Nova-Pak column filled with C18 4 μm silica gel, a Perkin-Elmer isocratic LC pump, a PE Nelson 900 series interface, a Spectra-Physics UV/Vis detector and a data station. The eluant (acetonitrile/methanol/0.02M ammonium acetate = 4: 1 :5) was run at 1.0 ml/min with UV detection at 228 nm. The retention times of DTPA-paclitaxel and paclitaxel were 1.38 and 8.83 min, respectively. Peak areas were quantitated and compared with standard curves to determine the DTPA-paclitaxel and paclitaxel concentrations. The estimated half-life of DTPA-paclitaxel in 0.5 M NaHCO3 solution is about 16 days at room temperature.
Effects of DTPA-paclitaxel on the growth of B 16 mouse melanoma cells in vitro:
Cells were seeded in 24-well plates at a concentration of 2.5 x 10 cells/ml and grown in a 50:50 Dulbecco's modified minimal essential medium (DEM) and F12 medium containing 10% bovine calf serum at 37°C for 24 hrs in a 97% humidified atmosphere of 5.5% CO2. The medium was then replaced with fresh medium containing paclitaxel or DTPA-paclitaxel in concentration ranging from 5 x 10"9 M to 75 x 10" M. After 40 hrs, the cells were released by trypsinization and counted in a Coulter counter. The final concentrations of DMSO (used to dissolve paclitaxel) and 0.05 M sodium bicarbonate solution (used to dissolve DTPA-paclitaxel) in the cell medium were less than 0.01%. This amount of solvent did not have any effect on cell growth as determined by control studies.
The effects of DTPA-paclitaxel on the growth of B16 melanoma cells are presented in FIG. 2. After a 40-h incubation with various concentrations, DTPA- paclitaxel and paclitaxel were compared as to cytotoxicity. The IC50 for paclitaxel and DTPA-paclitaxel are 15 nM and 7.5 nM, respectively. Antitumor effect on mammary carcinoma (MCa-4) tumor model:
Female C3Hf/Kam mice were inoculated with mammary carcinoma (MCa-4) in the muscles of the right thigh (5 x 10 cells/mouse). When the tumors had grown to 8 mm (approx. 2 wks), a single dose of paclitaxel or DTPA-paclitaxel was given at 10. 20 and 40 mg equivalent paclitaxel/kg body weight. In control studies, saline and absolute alcohol/Cremophor 50/50 diluted with saline (1 :4) were used. Tumor growth was determined daily, by measuring three orthogonal tumor diameters. When the tumor size reached 12 mm in diameter, the tumor growth delay was calculated. The mice were sacrificed when tumors were approximately 15 mm.
The tumor growth curve is shown in FIG. 3. Compared to controls, both paclitaxel and DTPA-paclitaxel showed antitumor effect at a dose of 40 mg/kg. The data were also analyzed to determine the mean number of days for the tumor to reach 12 mm in diameter. Statistical analysis showed that DTPA-paclitaxel delayed tumor growth significantly compared to the saline treated control at a dose of 40 mg/kg (p < 0.01). The mean time for the tumor to reach 12 mm in diameter was 12.1 days for DTPA-paclitaxel compared to 9.4 days for paclitaxel (FIG. 4).
Radiolabeling of DTPA-paclitaxel with In Into a 2-ml V-vial were added successively 40 μl 0.6 M sodium acetate (pH
5.3) buffer, 40 μl 0.06 M sodium citrate buffer (pH 5.5), 20 μl DTPA-paclitaxel solution in ethanol (2% w/v) and 20 μl InCl3 solution (1.0 mCi) in sodium acetate buffer (pH 5.5). After an incubation period of 30 min at room temperature, the labeled π ιIn-DTPA-paclitaxel was purified by passing the mixture through a C18 Sep-Pac cartridge using saline and subsequently ethanol as the mobile phase. Free 1 ' 'in-DTPA (<3%) was removed by saline, while In-DTPA-paclitaxel was collected in the ethanol wash. The ethanol was evaporated under nitrogen gas and the labeled product was reconstituted in saline. Radiochemical yield: 84%.
Analysis of ' ' ' In-DTPA-paclitaxel: HPLC was used to analyze the reaction mixture and purity of In-DTPA- paclitaxel. The system consisted of a LDC binary pump, a 100 x 8.0 mm (i.d.) Waters column filled with ODS 5 μm silica gel. The column was eluted at a flow rate of 1 ml/min with a gradient mixture of water and methanol (gradient from 0% to 85% methanol over 15 min). The gradient system was monitored with a Nal crystal detector and a Spectra-Physics UV Vis detector. As evidenced by HPLC analysis, purification by Sep-Pak cartridge removed most of the π In-DTPA, which had a retention time of 2.7 min. The ' In-DTPA was probably derived from traces of DTPA contaminant in the DTPA-paclitaxel. A radio-chromatogram of ' In-DTPA- paclitaxel correlated with its UV chromatogram, indicating that the peak at 12.3 min was indeed the target compound. Under the same chromatographic conditions, paclitaxel had a retention time of 17.1 min. The radiochemical purity of the final preparation was 90% as determined by HPLC analysis.
Whole-body scintigraphy:
Female C3Hf7Kam mice were inoculated with mammary carcinoma (MCa-4) in the muscles of the right thigh (5 x 105 cells). When the tumors had grown to 12 mm in diameter, the mice were divided into two groups. In group I, the mice were anesthetized by intraperitoneal injection of sodium pentobarbital, followed by In- DTPA-paclitaxel (100-200 mCi) via tail vein. A γ-camera equipped with a medium energy collimator was positioned over the mice (3 per group). A series of 5 min acquisitions were collected at 5, 30, 60, 120, 240 min and 24 hrs after injection. In group II, the same procedures were followed except that the mice were injected with
In-DTPA as a control. FIG. 5 shows gamma-scintigraphs of animals injected with "in-DTPA and '" In-DTPA-paclitaxel. π lIn-DTPA was characterized by rapid clearance from the plasma, rapid and high excretion in the urine with minimal retention in the kidney and negligible retention in the tumor, the liver, the intestine and other organs or body parts. In contrast, In-DTPA-paclitaxel exhibited a pharmacological profile resembling that of paclitaxel (Eiseman et al, 1994). Radioactivity in the brain was negligible. Liver and kidney had the greatest tissue:plasma ratios. Hepatobiliary excretion of radiolabeled DTPA-paclitaxel or its metabolites was one of the major routes for the clearance of the drug from the blood. Unlike paclitaxel, a significant amount of In-DTPA-paclitaxel was also excreted through kidney, which only played a minor role in the clearance of paclitaxel. The tumor had significant uptake of In-DTPA-paclitaxel. These results demonstrate that In-DTPA-paclitaxel is able to detect certain tumors and to quantify the uptake of In-DTPA-paclitaxel in the tumors, which in turn, may assist in the selection of patients for the paclitaxel treatment.
Example 2 Polyglutamic Acid-Paclitaxel
The present example demonstrates the conjugation of paclitaxel to a water- soluble polymer, poly (1-glutamic acid) (PG). The potential of water-soluble polymers used as drug carriers is well established (Kopecek, 1990; Maeda and Matsumura, 1989). In addition to its ability to solubilize otherwise insoluble drugs, the drug-polymer conjugate also acts as a slow-release depot for controlled drug release.
Synthesis of PG-Paclitaxel
PG was selected as a carrier for paclitaxel because it can be readily degraded by lysosomal enzymes, is stable in plasma and contains sufficient functional groups for drug attachment. Several antitumor drugs, including Adriamycin (Van Heeswijk et al, 1985; Hoes et al, 1985), cyclophosphamide (Hirano et al, 1979), and Ara-C
(Kato et al, 1984) have been conjugated to PG.
PG sodium salt (MW 34 K, Sigma, 0.35 g) was dissolved in water. The pH of the aqueous solution was adjusted to 2 using 0.2 M HCI. The precipitate was collected, dialyzed against distilled water, and lyophilized to yield 0.29 g PG.
To a solution of PG (75 mg, repeating unit FW 170, 0.44 mmol) in dry DMF (1.5 mL) was added 20 mg paclitaxel (0.023 mmol, molar ratio PG/paclitaxel = 19),
15 mg dicyclohexylcarbodiimide (DCC) (0.073 mmol) and a trace amount of dimethylaminopyridine (DMAP). The reaction was allowed to proceed at room temperature for 4 hrs. Thin" layer chromatography (TLC, silica) showed complete conversion of paclitaxel (Rf = 0.55) to polymer conjugate (Rf = 0, mobile phase, CHCl3/MeOH = 10:1). The reaction mixture was poured into chloroform. The resulting precipitate was collected and dried in a vacuum to yield 65 mg polymer-drug conjugate. By changing the weight ratio of paclitaxel to PG in the starting materials, polymeric conjugates of various paclitaxel concentrations can be synthesized.
The sodium salt of PG-paclitaxel conjugate was obtained by dissolving the product in 0.5 M NaHCO3. The aqueous solution of PG-paclitaxel was dialyzed against distilled water (MWCO 1 ,000) to remove low molecular weight contaminants and excess NaHCO3 salt. Lyophilization of the dialysate yielded 88.6 mg of white powder. The paclitaxel content in this polymeric conjugate was determined by UV (described below) as 21 % (w/w). Yield (conversion to polymer bound paclitaxel, UV): 93%. PG-paclitaxel with higher paclitaxel content (up to 35%) can be synthesized by this method by simply increasing the ratio of paclitaxel to PG used.
Η-NMR (GE model GN 500 spectrometer, 500 MHz, in D2O): δ = 7.75 to 7.36 ppm (aromatic components of paclitaxel); δ = 6.38 ppm (C10-H), 5.97 ppm (CI3- H), 5.63 and 4.78 ppm (C2'-H), 5.55-5.36 ppm (C3'-H and C2-H, m), 5.10 ppm (C5-H), 4.39 ppm (C7-H), 4.10 (C20-H), 1.97 ppm (OCOCH3), and 1.18-1.20 ppm (C-CH3) are assigned to aliphatic components of paclitaxel. Other resonances of paclitaxel were obscured by the resonances of PG. PG resonances at 4.27 ppm (H-α), 2.21 ppm (H- γ), and 2.04 ppm (H-β) are in accordance with pure PG spectrum. The couplings of polymer conjugated paclitaxel are too poorly resolved to be measured with sufficient accuracy. The solubility in water was > 20 mg paclitaxel/ml.
Characterization of PG-paclitaxel
Ultraviolet spectra (UV) were obtained on a Beckman DU-640 spectrophotometer (Fullerton, CA). The content of paclitaxel conjugated to PG was estimated by UV based on a standard curve generated with known concentrations of paclitaxel in methanol (λ = 228 nm), assuming that the polymer conjugate in water and the free drug in methanol had the same molar extinction coefficients and that both followed Lambert Beer's law. As shown by its UV spectrum, PG-paclitaxel has characteristic paclitaxel absorption with λ shifts from 228 to 230 nm. The concentration of paclitaxel in PG-paclitaxel was estimated based on standard curve generated with known concentrations of paclitaxel in methanol at absorption of 228 nm, assuming that the polymer conjugate in water at 230 nm and the free drug in methanol at 228 nm have the same molar extinction and both follow Lambert Beer's law.
Gel Permeation Chromatography Studies of PG- Paclitaxel
The relative molecular weight of PG-paclitaxel was characterized by gel permeation chromatography (GPC). The GPC system consisted of two LDC model III pumps coupled with LDC gradient master, a PL gel GPC column, and a Waters 990 photodiode array detector. The elutant (DMF) was run at 1.0 ml/min with UV detection set at 270 nm. Conjugation of paclitaxel to PG resulted in an increase in the molecular weight of PG-paclitaxel, as indicated by the shift of retention time from 6.4 min of PG to 5.0 min of PG-paclitaxel conjugate as analyzed by GPC. The calculated molecular weight of PG-paclitaxel containing 15-25% paclitaxel (w/w) is in the range of 45-55 kDa. The crude product contained a small molecular weight contaminant (retention time 8.0 to 10.0 min, and 11.3 min), which can be effectively removed by converting PG-paclitaxel to its sodium salt, followed by dialysis.
Hydrolytic degradation of PG-paclitaxel conjugate PG-paclitaxel was dissolved in phosphate-buffered solutions (PBS, 0.01 M) at pH 6.0, pH 7.4, and pH 9.6 at an equivalent paclitaxel concentration of 0.4 mM. The solutions were incubated at 37°C with gentle shaking. At selected time intervals, aliquots (100 μl) were removed, mixed with an equal volume of methanol and analyzed by high performance liquid chromatography (HPLC). The HPLC system consisted of a reversed-phase silica column (Nova-Pac, Waters, CA), a mobile phase of methanol-water (2:1, v/v) delivered at a flow rate of 1.0 ml min, and a photodiode detector. The concentration of PG bound paclitaxel, free paclitaxel and other degradation products in each sample was calculated by comparing the peak areas with a separately obtained standard curve prepared from paclitaxel, assuming that the molar extinction coefficient of each peak at 228 nm is the same as that of paclitaxel. The half-life of the conjugate, estimated to be 132, 40 and 4 days at pH 6.0, 7.4 and 9.6 respectively, were determined by a linear least-square regression analysis. HPLC analysis revealed that incubation of PG-paclitaxel in PBS solutions produced paclitaxel and several other species including one that is more hydrophobic than paclitaxel (metabolite- 1). In fact, the amount of metabolite- 1, which was most probably 7-epipaclitaxel, recovered in PBS at pH 7.4 surpassed that of paclitaxel after 100 hours of incubation (FIG. 6).
In vitro studies
Aliquots obtained from PBS solution at pH 7.4 were subjected to analysis by a tubulin polymerization assay. The tubulin assembly reaction was performed at 32°C in PEM buffer (pH 6.9) at a tubulin (bovine brain, Cytoskeleton Inc., Boulder, CO) concentration of 1 mg/ml (10 μM) in the presence of testing samples (1.0 μM equiv. Paclitaxel) and 1.0 mM GTP. Tubulin polymerization was followed by measuring the absorbance of the solution at 340 nm over time. After 15 min, calcium chloride (125 mM) was added to measure CaCl2-induced depolymerization of microtubules. While PG-paclitaxel freshly dissolved in PBS was inactive in producing microtubules, aliquots of PG-paclitaxel incubated for three days did result in tubulin polymerization. The microtubules formed were stable against CaCl2-induced depolymerization.
The effect of PG-paclitaxel on cell growth was also examined by tetrazolium salt (MTT) assay (Mosmann, 1983). MCF-7 cells or 13762F cells were seeded at 2 x 10 cells/ml in a 96-well microtiter plate treated 24 hrs later with various concentrations of PG-paclitaxel, paclitaxel or PG, and incubated for an additional 72 hr. MTT solution (20 μl, 5 mg/ml) was then added to each well and incubated for 4 hr. The supernatant was aspirated, and the MTT formazan formed by metabolically viable cells was measured by a microplate fluorescence reader at a wavelength of 590 nm. Over the three day period. PG-paclitaxel inhibited tumor cell proliferation to an extent similar to that of free paclitaxel. For human breast tumor cell line MCF-7, the resultant IC50 values were 0.59 μM for paclitaxel and 0.82 μM for PG-paclitaxel (measured in paclitaxel equivalent units). Against the 13762F cell line, sensitivity for PG-paclitaxel (IC50 = 1.86 μM) was comparable to that of paclitaxel (IC50 = 6.79 μM). For both cell lines, the IC50 of PG alone was greater than 100 μM.
In vivo antitumor activity
All animal work was carried out at the animal facility at M.D. Anderson Cancer Center in accordance with institutional guidelines. C3H/Kam mice were bred and maintained in a pathogen-free facility in the Department of Experimental Radiation Oncology. Solitary tumors were produced in the muscle of the right thigh of female C3H/Kam mice (25-30g) by injecting 5 x 10 murine ovarian carcinoma cells (OCa-I), mammary carcinoma (MCa-4), hepatocarcinoma (HCa-I) or fibrous sarcoma (FSa-II). In a parallel study, female Fischer 344 rats (125-150 g) were injected with 1.0 x 105 viable 13762F tumor cells in 0.1 ml PBS. Treatments were initiated when the tumors in mice had grown to 500 mm (10 mm in diameter), or when the tumors in rats had grown to 2400 mm (mean diameter 17 mm). A single dose of PG-paclitaxel in saline or paclitaxel in Cremophor EL vehicle was given in doses varying from 40 to 160 mg equiv. Paclitaxel/kg body weight. In control experiments, saline, Cremophor vehicle [50/50 Cremophor/ethanol diluted with saline (1 :4)], PG ( MW 38K) solution in saline, and a paclitaxel/PG mixture were used. Tumor growth was determined daily (FIG. 7A, 7B, 7C, 7D and 7E) by measuring three orthogonal tumor diameters. Tumor volume was calculated according to formula (A x B x C)/2. Absolute growth delay (AGD) in mice is defined as the time in days for tumors treated with various drugs to grow from 500 to 2,000 mm in mice minus the time in days for tumors treated with saline control to grow from 500 to 2,000 mm3. Table 1 summarizes acute toxicity of PG paclitaxel in rats in comparison with paclitaxel/Cremophor. Table 2 summarizes the data concerning the effect of PG- paclitaxel against MCa-4, FSa-II and HCa-I tumors in mice. The data are also summarized in FIG. 7A-FIG. 7E. Table 1 Acute Toxicity of PG-Paclitaxel in Fischer Rats*
Figure imgf000029_0001
Drugs were administered intravenously into 13762F tumor-bearing Fischer rats
(female, 130 g) in a single injection.
PG-paclitaxel solution was prepared by dissolving the conjugate in saline (8 mg equiv. paclitaxel/ml). The injected volume at 60 mg/kg was 0.975 ml per rat.
Paclitaxel Cremophor solution was prepared by dissolving paclitaxel in a 1 :1 mixture of ethyl alcohol and Cremophor (30 mg/ml). This stock solution was further diluted with saline (1 :4) before injection. The final concentration of paclitaxel in the solution was 6 mg/ml. The injected volume at 60 mg/kg was
1.3 ml per rat.
PG solution was prepared by dissolving the polymer in saline (22 mg/ml). The injected dose was 0.3 g/kg (1.8 ml per rat), which was equivalent to paclitaxel dose of 60 mg/kg.
Cremophor vehicle was prepared by diluting a mixture of ethyl alcohol and cremophor (1 :1) with saline (1 :4).
Table 2: The Antitumor Effect of PG-Paclitaxel Against Different Types of In Vivo Murine Tumors
Tumor Drug" Time to Grow00 AGDC estα 500 - 2000 mm3
MCa-4 Saline 4.8±0.8 (5) - -
PG (0.6 g/kg) 9.3+1.1 (4) 4.5 0.0114
Cremophor Vehicle 6.1±0.7 (5) 1.3 0.265
PG-Pacl (40 mg/kg) 8.6±1.2 (4) 3.8 0.026
PG-Pacl (60 mg/kg) 14.2+1.1 (5) 9.4 0.0001
PG-Pacl (120 mg/kg) 44.4±2.9 (5) 39.6 O.0001
Paclitaxel (40 mg/kg) 9.0+0.6 (4) 4.2 0.0044
Paclitaxel (60 mg/kg) 9.3±0.3 (5) 4.5 0.0006
FSa-II Saline 1.9±0.1 (5) - .
PG (0.8 g/kg) 2.8±0.2 (6) 0.9 0.0043
Cremophor Vehicle 2.2+0.2 (6) 0.3 0.122
PG-Pacl (80 mg/kg) 3.8+0.4 (6) 1.9 0.0016
PG-Pacl (160 mg/kg) 5.1±0.3 (13) 3.2 O.0001
Paclitaxel (80 mg/kg) 4.2±0.3 (6) 2.3 0.0002
PG+Paclitaxel 3.0±0.2 (6) 1.1 0.0008
HCa-I Saline 7.3±0.3 (5) - -
PG (0.8 g/kg) 7.7±0.4 (4) 0.4 0.417
Cremophor Vehicle 6.8±0.8 (5) -0.5 0.539
PG-Pacl (40 mg kg) 8.2±0.7 (5) 0.9 0.218
PG-Pacl (80 mg/kg) 8.6±0.2 (5) 1.3 0.0053
PG-Pacl (160 mg/kg) 11.0±0.8 (4) 3.7 0.0023
Paclitaxel (80 mg/kg) 6.4±0.5 (5) -0.9 0.138
PG+Paclitaxel 6.7±0.4 (5) -0.6 0.294
a Mice bearing 500 mm tumors in the right leg were treated with various doses of
PG-paclitaxel (40-120 mg equiv. paclitaxel/kg) in saline or paclitaxel in Cremophor vehicle i.v. in a single injection. Control animals were treated with saline (0.6 ml), Cremophor vehicle (0.5 ml), PG solution in saline, or PG g/kg) plus paclitaxel (80 mg/kg). b Tumor growth was determined by daily measurement of three orthogonal diameters with calipers and the volume was calculated as (a x b x c)/2. Shown in brackets are the number of mice used in each group. The time in days to grow from 500 mm to 2000 mm are presented mean±standard deviation. c Absolute growth delay (AGD) defined as the time in days for tumors treated with various drugs to grow from 500 to 2000 mm3 minus the time in days for tumors treated with saline control to grow from 500 to 2000 mm3, d The time in days to grow from 500 to 2000 mm were compared for treatment groups and saline group using Student's /-Test. P- values are two-sided and were taken to be significant when less than to equal 0.05. Example 3 Polyethylene glycol-Paclitaxel
Synthesis of polyethylene glycol-paclitaxel (PEG-paclitaxel) The synthesis was accomplished in two steps. First 2'-succinyl-paclitaxel was prepared according to a reported procedure (Deutsch et al, 1989). Paclitaxel (200 mg, 0.23 mmol) and succinic anhydride (288 mg, 2.22 mmol) were allowed to react in anhydrous pyridine (6 ml) at room temperature for 3 hrs. The pyridine was then evaporated, and the residue was treated with water, stirred for 20 min, and filtered. The precipitate was dissolved in acetone, water was slowly added, and the fine crystals were collected to yield 180 mg 2'-succinyl-paclitaxel. PEG-paclitaxel was synthesized by an N-ethoxycarbonyl-2-ethoxy-l,2-dihydroquinoline (EEDQ) mediated coupling reaction. To a solution of 2'-succinyl-paclitaxel (160 mg, 0.18 mmol) and methoxypolyoxyethylene amine (PEG-NH2, MW 5000, 900 mg, 0.18 mmol) in methylene chloride was added EEDQ (180 mg, 0.72 mmol). The reaction mixture was stirred at room temperature for 4 hrs. The crude product was chromatographed on silica gel with ethyl acetate followed by chloroform-methanol (10:1). This gave 350 mg of product. ]H NMR (CDC13) δ 2.76 (m, succinic acid, COCH2CH2CO2), δ 3.63 (PEG, OCH2CH2O), δ 4.42 (C7-H) and δ 5.51 (C2'-H). Maximal UV absorption was at 288 nm which is also characteristic for paclitaxel. Attachment to PEG greatly improved the aqueous solubility of paclitaxel (>20 mg equivalent paclitaxel/ml water).
Hydrolytic stability of PEG-Paclitaxel PEG-Paclitaxel was dissolved in phosphate buffer (0.01M) at various pHs at a concentration of 0.4 mM and the solutions were allowed to incubate at 37°C with gentle shaking. At selected time intervals, aliquots (200 μl) were removed and lyophilized. The resulting dry powders were redissolved in methylene chloride for gel permeation chromatography (GPC analysis). The GPC system consisted of a Perkin- Elmer PL gel mixed bed column, a Perkin-Elmer isocratic LC pump, a PE Nelson 900 series interface, a Spectra-Physics UV/Vis detector and a data station. The elutant (methylene chloride) was run at 1.0 ml/min with the UV detector set at 228 nm. The retention times of PEG-paclitaxel and paclitaxel were 6.1 and 8.2 min, respectively. Peak areas were quantified and the percentage of PEG-paclitaxel remaining and the percentage of paclitaxel released were calculated. The half life of PEG-paclitaxel determined by linear least-squares at pH 7.4 was 54 min. The half-life at pH 9.0 was 7.6 min. Release profiles of paclitaxel from PEG-paclitaxel at pH 7.4 is shown in FIG. 8.
Cytotoxicity Studies of PEG-paclitaxel using B16 mouse melanoma cells in vitro Following the procedure described in the cytotoxicity studies with DTPA- paclitaxel, melanoma cells were seeded in 24-well plates at a concentration of 2.5 x 10 cells/ml and grown in a 50:50 Dulbecco's modified minimal essential medium (DME) and F12 medium containing 10% bovine calf serum at 37°C for 24 hrs in a 97% humidified atmosphere of 5.5% CO2. The medium was then replaced with fresh medium containing paclitaxel or its derivatives in concentrations ranging from 5 x 10" 9 M to 75 x 10"9 M. After 40 hrs, the cells were released by trypsinization and counted in a Coulter counter. The final concentrations of DMSO (used to dissolve paclitaxel) and 0.05 M sodium bicarbonate solution (used to dissolve PEG-paclitaxel) in the cell medium were less than 0.01%. This amount of solvent did not have any effect on cell growth as determined by control studies. Furthermore, PEG in the concentration range used to generate an equivalent paclitaxel concentration from 5 x 10"9 M to 75 x 10"9 M also did not effect cell proliferation.
Antitumor effect of PEG-paclitaxel against MCa-4 tumor in mice To evaluate the antitumor efficacy of PEG-paclitaxel against solid breast tumors, MCa-4 cells (5 x 10s cells) were injected into the right thigh muscle of female C3Hf/Kam mice. As described in Example 1 with the DTPA-paclitaxel, when the tumors were grown to 8 mm (Approx. 2 wks), a single dose of paclitaxel or PEG- paclitaxel was given at 10, 20 and at 40 mg equivalent paclitaxel/kg body weight. Paclitaxel was initially dissolved in absolute ethanol with an equal volume of Cremophor. This stock solution was further diluted (1 :4 by volume) with a sterile physiological solution within 15 minutes of injection. PEG-paclitaxel was dissolved in saline (6 mg equiv. paclitaxel/ml) and filtered through a sterile filter (Millipore, 4.5 μm). Saline, paclitaxel vehicle, absolute alcohol: Cremophor (1 : 1) diluted with saline (1 :4) and PEG solution in saline (600 mg/kg body weight) were used in control experiments. Tumor growth was determined daily, by measuring three orthogonal tumor diameters. When the tumor size reached 12 mm in diameter, the tumor growth delay was calculated.
The tumor growth curve is shown in FIG 9. At a dose of 40 mg/kg, both PEG- paclitaxel and paclitaxel effectively delayed tumor growth. Paclitaxel was more effective than PEG-paclitaxel, although the difference was not statistically significant. Paclitaxel treated tumors required 9.4 days to reach 12 mm in diameter whereas PEG- paclitaxel-treated tumors required 8.5 days. Statistically, these values were significant (p > 0.05) as compared to their corresponding controls, which were 6.7 days for the paclitaxel vehicle and 6.5 days for the saline solution of PEG (FIG. 4).
While the compositions and methods of this invention have been described in terms of preferred embodiments, it will be apparent to those of skill in the art that variations may be applied to the compositions, methods and in the steps or in the sequence of steps of the methods described herein without departing from the concept, spirit and scope of the invention. More specifically, it will be apparent that certain agents which are both chemically and physiologically related may be substituted for the agents described herein while the same or similar results would be achieved. All such similar substitutes and modifications apparent to those skilled in the art are deemed to be within the spirit, scope and concept of the invention as defined by the appended claims. References
The following references, to the extent that they provide exemplary procedural or other details supplementary to those set forth herein, are specifically incorporated herein by reference.
Bartoni and Boitard, "In vitro and in vivo antitumoral activity of free, and encapsulated taxol," J Microencapsulation, 7:191-197, 1990.
Cortes, J.E. and Pazdur, R., "Docetaxel", Journal of Clinical Oncology, 13:2643- 2655, 1995.
Deutsch et al, "Synthesis of congeners and prodrugs. 3. water-soluble prodrugs of taxol with potent antitumor activity," J. Med. Chem., 32:788-792, 1989.
Eiseman et al, "Plasma pharmacokinetics and tissue distribution of paclitaxel in CD2F1 mice," Cancer Chemother. Pharmacol, 34:465-471, 1994.
Fidler, et al, "The biology of cancer invasion and metastasis," Adv. Cancer Res., 28: 149-250, 1987.
Goldspiel, "Taxol pharmaceutical issues: preparation, administration, stability, and compatibility with other medications," Ann. Pharmacotherapy, 28:S23-26, 1994.
Greenwald et al, "Highly water soluble Taxol derivatives, 7-polyethylene glycol esters as potential products," J. Org. Chem., 60:331-336, 1995.
Greenwald et al, "Highly water soluble taxol derivative: 2'-polyethylene glycol esters as potential products," Bioorganic & Medicinal Chemistry Letters, 4:2465-2470, 1994.
Hirano et al. , "Polymeric derivatives of activated cyclophosphamide as drug delivery systems in antitumor therapy," Makromol Chem., 180:1125-1130, 1979. Hoes et al, "Optimization of macromolecular prodrugs of the antitumor antibiotic adriamycin," J. Controlled Release, 2:205-213, 1985.
Hofle e/ α/., DE38942.
Horwitz et al. , "Taxol, mechanisms of action and resistance," J. Natl. Cancer Inst. Monographs No. 15, pp. 55-61, 1993. Kato et al, "Antitumor activity of 1 -b-arabinofuranosylcytosine conjugated with polyglutamic acid and its derivative", Cancer Res., 44:25, 1984.
Kopecek, "The potential of water-soluble polymeric carriers in targeted and site- specific drug delivery", J. Controlled Release, 11 :279-290, 1990.
Kopecek and Kopeckova, "Targetable water-soluble polymeric anticancer drugs: achievements and unsolved problems," Proceed. Intern Symp. Control. Rel. Bioact. Mater., 20:190-191, 1993.
Maeda and Matsumura, "Tumoritropic and lymphotropic principles of macromolecular drugs", Critical Review in Therapeutic Drug Carrier Systems, 6:193- 210, 1989.
Magri and Kingston, "Modified taxols. 2. Oxidation products of taxol," J. Org. Chem., 51 :797-802, 1986.
Mathew et al, "Synthesis and evaluation of some water-soluble prodrugs and derivatives of taxol with antitumor activity," J. Med. Chem., 35:145-151, 1992.
Mosmann, T„ "Rapid colormetric assay for cellular growth and survival: application to proliferation and cytotoxic assay," J. Immunol. Methods, 65:55-63, 1983.
Oliver, S . et al, Suppression of collagen-induced arthritis using an angiogenesis inhibitor, AGM-1470, and a microtubule stabilizer, Taxol," Cellular Immunology, 157:291-299, 1994.
Phillips-Hughes and Kandarpa, "Restenosis: pathophysiology and preventive strategies," JVIR, 7:321-333, 1996.
Reynolds, T., "Polymers help guide cancer drugs to tumor targets- and keep them there," J. Natl. Cancer Institute, 87:1582-1584, 1995.
Scudiero et al. "Evaluation of a Soluble Tetrazolium/Formazan Assay for Cell Growth and Drug Sensitivity in Culture Using Human and Other Tumor Cell Lines," Cancer Research, 48:4827-4833, 1988.
Serruys et al. , "A comparison of balloon-expandable-stent implantation with balloon angioplasty in patients with coronary artery disease," N. Engl. J. Med., 331:489-495, 1994.
Sharma and Straubinger, "Novel taxol formulations: Preparation and Characterization of taxol-containing liposomes," Pharm. Res., 11 :889-896, 1994.
US Patent 5,583,153 van Heeswijk et al, "The synthesis and characterization of polypeptide-adriamycin conjugate and its complexes with adriamycin. Part 1 ", J. Controlled Release, 1:301- 315, 1985.
Weiss et al , "Hypersensitivity reactions from Taxol," J. Clin. Oncol, 8:1263-1268, 1990.
WO 96/25176
Zhao, Z. and Kingston, D.G.I. , "Modified taxols. 6. Preparation of water-soluble taxol phosphates," J. Nat. Prod, 54:1607-1611, 1991.

Claims

CLAIMS:
1. A composition comprising an anti-tumor drug conjugated to a water soluble polymer or metal chelator and wherein said anti-tumor drug is paclitaxel. docetaxel, etopside, teniposide, camptothecin or epothilone.
The composition of claim 1 wherein the anti-tumor drug is paclitaxel.
3. The composition of claim 1, wherein said anti-tumor drug is docetaxel.
4. The composition of claim 1, wherein said anti-tumor drug is conjugated to a water soluble metal chelator.
5. The composition of claim 4, further comprising a chelated metal ion.
6. The composition of claim 5, wherein said chelated metal ion is selected from the group consisting of aluminum, boron, calcium, chromium, cobalt, copper, dysprosium, erbium, europium, gadolinium, gallium, germanium, holmium, indium, iridium, iron, magnesium, manganese, nickel, platinum, rhenium, rubidium, ruthenium, samarium, sodium, technetium, thallium, tin, yttrium and zinc.
The composition of claim 5, wherein said chelated metal ion is a radionuclide.
8. The composition of claim 7, wherein said radionuclide is selected from the group consi s *ting o ff blr G-a, 68 G^a, H I I in, 99mτ Tc, 90v Y, l l mτ In and , 1931^ P*t.
9. A composition comprising In-DTPA-paclitaxel.
10. The composition of claim 4, wherein said water soluble chelator is selected from the group consisting of diethylenetriaminepentaacetic acid (DTPA), ethylenediaminetetraacetic acid (EDTA), l,4,7,10-tetraazacyclododecane-N,N',N" ,N'"-tetraacetate (DOTA), tetraazacyclotetradecane-N,N',N"N",-tetraacetic acid (TETA), hydroxyethylidene diphosphonate (HEDP), dimercaptosuccinic acid (DMSA), diethylenetriaminetetramethylenephosphonic acid (DTTP), DPDP and l-(p- aminobenzyl)-DTPA.
11. The composition of claim 4, wherein said chelator is diethylenetriaminepentaacetic acid (DTPA).
12. The composition of claim 1, wherein said water soluble polymer is selected from the group consisting of poly (d-glutamic acid), poly (1-glutamic acid), poly (dl- glutamic acid), poly (d-aspartic acid), poly (1-aspartic acid), poly (dl-aspartic acid), polyethylene glycol, polyacrylic acid, poly(2-hydroxyethyl 1-glutamine), carboxymethyl dextran, hyaluronic acid, human serum albumin, alginic acid and a combination thereof.
13. The composition of claim 12, wherein said polymer is further defined as a copolymer with polycaprolactone, polyglycolic acid, polylactic acid, polyacrylic acid, poly(2-hydroxyethyl 1-glutamine), carboxymethyl dextran, hyaluronic acid, human serum albumin, polyalginic acid or a combination thereof.
14. The composition of claim 12. wherein said polymer has a molecular weight of from about 5,000 to about 100,000.
15. The composition of claim 12, wherein said polymer has a molecular weight of from about 20,000 to about 80,000.
16. The composition of claim 12, wherein said polymer has a molecular weight of about 30,000 to about 60,000.
17. The composition of claim 12, wherein said water soluble polymer is conjugated to the 2'-and/or the 7-hydroxyl of paclitaxel or docetaxel.
18. The composition of claim 12, wherein said water soluble polymer is polyethylene glycol.
19. The composition of claim 12, wherein said water soluble polymer is poly (1- glutamic acid).
20. The composition of claim 12, wherein said water soluble polymer is poly (1- aspartic acid).
21. The composition of claim 1 dispersed in a pharmaceutically acceptable carrier solution.
22 A method of determining anti-tumor drug uptake by tumor tissue comprising the steps of:
a) obtaining a composition comprising a conjugate of paclitaxel or docetaxel and a metal chelator and a chelated metal ion;
b) contacting said tumor tissue with said composition; and
c) detecting the presence of said chelated metal ion in said tumor tissue;
wherein the presence of said chelated metal ion in said tumor tissue is indicative of paclitaxel by said tumor tissue.
23. A method of claim 22 wherein the anti-tumor drug is paclitaxel.
24. The method of claim 22, wherein said chelated metal ion is a radionuclide and said detection is scintigraphic.
25. The method of claim 22, wherein said tumor tissue is in a subject and said composition is administered to said subject.
26. A method of treating cancer in a subject comprising the steps of: a) obtaining a composition comprising paclitaxel or docetaxel conjugated to a water soluble polymer or chelator and dispersed in a pharmaceutically acceptable solution; and
b) administering said solution to said subject in an amount effective to treat said cancer.
27. The method of claim 26 wherein said composition comprises paclitaxel.
28. The method of claim 26, further comprising determining the paclitaxel or docetaxel uptake of said tumor prior to said administering, wherein said determining is by administering a chelator-metal ion-paclitaxel or docetaxel conjugate to said subject and detecting the presence of said metal ion in said tumor.
29. The method of claim 26, wherein said cancer is breast cancer, ovarian cancer, malignant melanoma, lung cancer, gastric cancer, colon cancer, head and neck cancer or leukemia.
30. The method of claim 26, wherein said cancer is breast cancer.
31. The method of claim 26, wherein said cancer is ovarian cancer.
32. A method of decreasing at least one symptom of a systemic autoimmune disease comprising administering to a subject having a systemic autoimmune disease an effective amount of a composition comprising paclitaxel or docetaxel conjugated to poly (1-glutamic acid) or poly (1-aspartic acid).
33. The method of claim 32, wherein said composition comprises paclitaxel.
34. The method of claim 32, wherein said composition comprises poly (1-glutamic acid).
35. The method of claim 32, wherein said system autoimmune disease is rheumatoid arthritis.
36. A method of inhibiting arterial restenosis or arterial occlusion following vascular trauma comprising administering to a subject in need thereof, a composition comprising paclitaxel or docetaxel conjugated to poly (1-glutamic acid) or poly (1- aspartic acid).
37. The method of claim 36, wherein said composition comprises paclitaxel.
38. The method of claim 36, wherein said composition comprises poly (1-glutamic acid).
39. The method of claim 36 wherein said subject is a coronary bypass, vascular surgery, organ transplant or coronary or arterial angioplasty patient.
40. The method of claim 36, wherein said composition is coated on a stent and said stent is implanted at the sight of vascular trauma.
41. A pharmaceutical composition comprising paclitaxel or docetaxel conjugated to a water soluble polymer or chelator.
42. The composition of claim 41, wherein said water soluble polymer is poly (1- aspartic acid) or poly (1-glutamic acid).
43. The composition of claim 41 , wherein said chelator is DTPA.
44. The composition of claim 43, further comprising a therapeutic amount of a chelated radionuclide.
45. A method of obtaining a body image of a subject comprising:
a) administering to said subject an effective amount of the agent of claim 6; and
b) measuring scintigraphic signals to obtain an image.
46. An implantable medical device, wherein said device is coated with a composition comprising paclitaxel or docetaxel conjugated to polyglutamic acid or polyaspartic acid in an amount effective to inhibit smooth muscle cell proliferation.
47. The implantable medical device of claim 46, further defined as a stent coated with said composition.
48. The implantable medical device of claim 47, wherein said stent is adapted to be used after balloon angioplasty and said composition is effective to inhibit restenosis.
49. A composition comprising polyglutamic acid conjugated to the 2' or 7 hydroxyl of paclitaxel.
50. A composition comprising polyaspartic acid conjugated to the 2' or 7 hydroxyl of paclitaxel.
51. The composition of claim 49, wherein said polyglutamic acid is conjugated to the 2' and the 7 hydroxyl of paclitaxel.
52. The composition of claim 50, wherein said polyaspartic acid is conjugated to the 2' and the 7 hydroxyl of paclitaxel.
PCT/US1997/003687 1996-03-12 1997-03-11 Water soluble paclitaxel prodrugs WO1997033552A1 (en)

Priority Applications (16)

Application Number Priority Date Filing Date Title
DE69735057T DE69735057T2 (en) 1996-03-12 1997-03-11 WATER-SOLUBLE PACLITAXEL PRODUCTS
KR1019980711024A KR100561788B1 (en) 1996-03-12 1997-03-11 Compositions comprising water-soluble paclitaxel prodrugs and implantable medical devices comprising such compositions
CA002250295A CA2250295C (en) 1996-03-12 1997-03-11 Water soluble paclitaxel prodrugs
HU9903952A HU226646B1 (en) 1996-03-12 1997-03-11 Water soluble pharmaceutical compositions containing taxane derivatives
JP53273497A JP3737518B2 (en) 1996-03-12 1997-03-11 Water-soluble paclitaxel prodrug
PL97328807A PL189698B1 (en) 1996-03-12 1997-03-11 Water-soluble promedicines of paklitaxel
EP97917512A EP0932399B1 (en) 1996-03-12 1997-03-11 Water soluble paclitaxel prodrugs
EA199800817A EA002400B1 (en) 1996-03-12 1997-03-11 Water soluable prodrugs of anti-tumor action
IL12617997A IL126179A (en) 1996-03-12 1997-03-11 Pharmaceutical compositions containing anti-tumor drug conjugates
BR9710646-1A BR9710646A (en) 1996-03-12 1997-03-11 Water soluble prodrugs of paclitaxel.
NZ332234A NZ332234A (en) 1996-03-12 1997-03-11 Water soluble paclitaxel prodrugs formed by conjugating paclitaxel or docetaxel with a polyglutamic acid polymer and use for treating cancer
SI9730728T SI0932399T1 (en) 1996-03-12 1997-03-11 Water soluble paclitaxel prodrugs
AU25806/97A AU735900B2 (en) 1996-03-12 1997-03-11 Water soluble paclitaxel prodrugs
UA98105316A UA68330C2 (en) 1996-03-12 1997-11-03 Water soluble anticancer pharmaceutical composition (variants) and method for treatment (variants)
NO19984210A NO324461B1 (en) 1996-03-12 1998-09-11 A composition comprising an anti-tumor drug conjugated to a water-soluble polymer, the use thereof and an implantable medical device comprising the composition
NO20072562A NO332539B1 (en) 1996-03-12 2007-05-21 Use of a mixture of a water-soluble anti-tumor drug and a platinum drug for the manufacture of a drug preparation for the treatment of disease

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US1318496P 1996-03-12 1996-03-12
US60/013,184 1996-03-12

Publications (1)

Publication Number Publication Date
WO1997033552A1 true WO1997033552A1 (en) 1997-09-18

Family

ID=21758713

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1997/003687 WO1997033552A1 (en) 1996-03-12 1997-03-11 Water soluble paclitaxel prodrugs

Country Status (23)

Country Link
US (2) US5977163A (en)
EP (2) EP1683520B1 (en)
JP (2) JP3737518B2 (en)
KR (1) KR100561788B1 (en)
CN (2) CN1304058C (en)
AT (1) ATE314843T1 (en)
AU (1) AU735900B2 (en)
BR (1) BR9710646A (en)
CA (1) CA2250295C (en)
CZ (1) CZ297979B6 (en)
DE (1) DE69735057T2 (en)
DK (1) DK0932399T3 (en)
EA (1) EA002400B1 (en)
ES (2) ES2448467T3 (en)
HU (1) HU226646B1 (en)
IL (1) IL126179A (en)
NO (2) NO324461B1 (en)
NZ (1) NZ332234A (en)
PL (1) PL189698B1 (en)
PT (1) PT932399E (en)
SI (1) SI0932399T1 (en)
UA (1) UA68330C2 (en)
WO (1) WO1997033552A1 (en)

Cited By (116)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1997040854A2 (en) * 1996-05-01 1997-11-06 Antivirals Inc. Polypeptide conjugates for transporting substances across cell membranes
EP0895784A1 (en) * 1996-04-15 1999-02-10 Asahi Kasei Kogyo Kabushiki Kaisha Medicament composite
WO1999016416A2 (en) * 1997-09-29 1999-04-08 Schering Ag Coated medical implants, method for producing them, and use of the same for preventing restenosis
WO1999033473A1 (en) * 1997-12-25 1999-07-08 Toray Industries, Inc. Remedies for intramedullary diseases
WO1999039694A2 (en) * 1998-02-05 1999-08-12 Novartis Ag Compositions containing organic compounds
US5977163A (en) * 1996-03-12 1999-11-02 Pg-Txl Company, L. P. Water soluble paclitaxel prodrugs
WO2000000238A1 (en) * 1998-06-26 2000-01-06 Quanam Medical Corporation Topoisomerase inhibitors for prevention of restenosis
WO2000001682A1 (en) * 1998-07-01 2000-01-13 Bcm Developpement Inc. Water-soluble derivatives of paclitaxel, method for producing same and uses thereof
WO2000003677A2 (en) * 1998-07-14 2000-01-27 Yissum Research Development Company Of The Hebrew University Of Jerusalem Use of bisphosphonates in the treatment of vascular restenosis
US6030941A (en) * 1996-05-01 2000-02-29 Avi Biopharma, Inc. Polymer composition for delivering substances in living organisms
WO2000010622A1 (en) * 1998-08-20 2000-03-02 Cook Incorporated Coated implantable medical device
WO2000018439A2 (en) * 1998-09-29 2000-04-06 Schering Aktiengesellschaft Use of neoangiogenesis markers for diagnosing and treating tumours by therapy
WO2000044443A2 (en) * 1999-01-29 2000-08-03 Angiotech Pharmaceuticals Inc. Intra-pericardial delivery of anti-microtubule agents
EP1028756A1 (en) * 1998-03-30 2000-08-23 PG-TXL Company, L.P. Water soluble paclitaxel derivatives
WO2000050016A2 (en) * 1999-02-23 2000-08-31 Angiotech Pharmaceuticals, Inc. Compositions and methods for improving integrity of compromised body passageways and cavities
US6179817B1 (en) 1995-02-22 2001-01-30 Boston Scientific Corporation Hybrid coating for medical devices
WO2001010342A1 (en) * 1999-08-10 2001-02-15 Scimed Life Systems, Inc. Thrombosis filter having a surface treatment
WO2001019406A2 (en) * 1999-09-13 2001-03-22 Nobex Corporation Amphiphilic prodrugs
WO2001026693A2 (en) * 1999-10-12 2001-04-19 Cell Therapeutics, Inc. Manufacture of polyglutamate-therapeutic agent conjugates
WO2001070275A2 (en) * 2000-03-17 2001-09-27 Cell Therapeutics, Inc. Polyglutamic acid-camptothecin conjugates and methods of preparation
US6306993B1 (en) 1997-05-21 2001-10-23 The Board Of Trustees Of The Leland Stanford, Jr. University Method and composition for enhancing transport across biological membranes
JP2001526224A (en) * 1997-12-22 2001-12-18 シェーリング コーポレイション Combinations of benzocycloheptapyridine compounds and antitumor agents for treating proliferative diseases
JP2002511845A (en) * 1997-04-24 2002-04-16 ニユコメド・イメージング・アクシエセルカペト Contrast agent
US6380405B1 (en) 1999-09-13 2002-04-30 Nobex Corporation Taxane prodrugs
WO2002058688A1 (en) * 2001-01-24 2002-08-01 Mestex Ag Use of neurotoxic substances in producing a medicament for treating joint pains
US6458373B1 (en) 1997-01-07 2002-10-01 Sonus Pharmaceuticals, Inc. Emulsion vehicle for poorly soluble drugs
JP2002534461A (en) * 1999-01-12 2002-10-15 クアナム メディカル コーポレイション Compositions and methods for administration of water-insoluble paclitaxel derivatives
EP1251739A2 (en) * 1999-04-13 2002-10-30 Fannin Bioscience, Inc. Poly(dipeptide) as a drug carrier
US6541508B2 (en) 1999-09-13 2003-04-01 Nobex Corporation Taxane prodrugs
EP1301500A1 (en) * 2000-06-22 2003-04-16 Nitromed, Inc. Nitrosated and nitrosylated taxanes, compositions and methods of use
WO2003037385A1 (en) * 2001-10-30 2003-05-08 Nektar Therapeutics Al, Corporation Water-soluble polymer conjugates of retinoic acid
US6593292B1 (en) 1999-08-24 2003-07-15 Cellgate, Inc. Compositions and methods for enhancing drug delivery across and into epithelial tissues
US6638906B1 (en) 1999-12-13 2003-10-28 Nobex Corporation Amphiphilic polymers and polypeptide conjugates comprising same
US6669951B2 (en) 1999-08-24 2003-12-30 Cellgate, Inc. Compositions and methods for enhancing drug delivery across and into epithelial tissues
US6683100B2 (en) 1999-01-19 2004-01-27 Novartis Ag Organic compounds
WO2004035629A2 (en) * 2002-10-18 2004-04-29 Fidia Farmaceutici S.P.A. Taxanes covalently bounded to hyaluronic acid or hyaluronic acid derivatives
US6730293B1 (en) 1999-08-24 2004-05-04 Cellgate, Inc. Compositions and methods for treating inflammatory diseases of the skin
EP1475105A1 (en) * 2003-05-09 2004-11-10 Schering AG Bone localising radiopharmaceutical and tubulin-interacting compound combinatorial radiotherapy
EP1478406A1 (en) * 2002-02-27 2004-11-24 Pharmain, Ltd. Compositions for delivery of therapeutics and other materials, and methods of making and using the same
EP1561436A1 (en) * 2004-02-05 2005-08-10 SORIN BIOMEDICA CARDIO S.r.l. A stent for endoluminal delivery of active principles or agents
WO2005094897A2 (en) * 2004-04-01 2005-10-13 Cardiome Pharma Corp. Pegylated ion channel modulating compounds
US6984400B2 (en) 1998-07-14 2006-01-10 Yissum Research Development Company Of The Hebrew University Of Jerusalem Method of treating restenosis using bisphosphonate nanoparticles
US7008645B2 (en) 1998-07-14 2006-03-07 Yissum Research Development Company Of The Hebrew University Of Jerusalem Method of inhibiting restenosis using bisphosphonates
US7030155B2 (en) 1998-06-05 2006-04-18 Sonus Pharmaceuticals, Inc. Emulsion vehicle for poorly soluble drugs
US7067111B1 (en) 1999-10-25 2006-06-27 Board Of Regents, University Of Texas System Ethylenedicysteine (EC)-drug conjugates, compositions and methods for tissue specific disease imaging
US7153864B2 (en) 2000-03-17 2006-12-26 Cell Therapeutics Inc. Polyglutamic acid-camptothecin conjugates and methods of preparation
WO2007027941A3 (en) * 2005-08-31 2007-04-26 Abraxis Bioscience Inc Compositions and methods for preparation of poorly water soluble drugs with increased stability
US7229961B2 (en) 1999-08-24 2007-06-12 Cellgate, Inc. Compositions and methods for enhancing drug delivery across and into ocular tissues
JP2007153905A (en) * 1999-12-16 2007-06-21 F Hoffmann La Roche Ag Substituted bisindolylmaleimide for inhibition of cell proliferation
EP1800702A2 (en) * 1998-10-14 2007-06-27 Boston Scientific Limited Loading and release of water-insoluble drugs
EP1800664A1 (en) * 1998-02-05 2007-06-27 Novartis AG Pharmaceutical formulations for parenteral administration comprising epothilone
US7261875B2 (en) 2001-12-21 2007-08-28 Board Of Regents, The University Of Texas System Dendritic poly (amino acid) carriers and methods of use
WO2007124700A3 (en) * 2006-05-03 2007-12-21 I Q A A S Pharmaceutical composition containing taxane derivative destined for the preparation of an infusion solution, method of preparation thereof and use thereof
AU2006222697B2 (en) * 2001-03-26 2008-03-13 Bayer Intellectual Property Gmbh Preparation for the Prophylaxis of Restenosis
WO2008062945A1 (en) * 2006-11-22 2008-05-29 Standard Sci-Tech Inc. Stent
WO2007015761A3 (en) * 2005-07-21 2009-04-16 Fmc Biopolymer As Medical devices coated with a fast dissolving biocompatible coating
US7705036B2 (en) 2004-04-01 2010-04-27 Cardiome Pharma Corp. Deuterated aminocyclohexyl ether compounds and processes for preparing same
US7744861B2 (en) 2003-09-17 2010-06-29 Nektar Therapeutics Multi-arm polymer prodrugs
US7754897B2 (en) 2005-06-15 2010-07-13 Cardiome Pharma Corp. Synthetic processes for the preparation of aminocyclohexyl ether compounds
US7846445B2 (en) 2005-09-27 2010-12-07 Amunix Operating, Inc. Methods for production of unstructured recombinant polymers and uses thereof
US7855279B2 (en) 2005-09-27 2010-12-21 Amunix Operating, Inc. Unstructured recombinant polymers and uses thereof
US7932377B2 (en) 2003-05-30 2011-04-26 Supramol Parenteral Colloids Gmbh Complexing of medicinal substances with high-molecular carriers and injection and infusion solutions containing said complexes
US7960336B2 (en) 2007-08-03 2011-06-14 Pharmain Corporation Composition for long-acting peptide analogs
US8101164B2 (en) 2003-09-05 2012-01-24 The General Hospital Corporation Dual phase drug release system
RU2447443C2 (en) * 2007-03-06 2012-04-10 Селл Терапьютикс Инк. Method for determining amount of conjugated taxane in polyglutamic acid-taxane conjugates
US8231859B2 (en) 2002-02-27 2012-07-31 Pharmain Corporation Compositions for delivery of therapeutics and other materials
US8329853B2 (en) 2006-06-15 2012-12-11 Cell Therapeutics, Inc. Process for the preparation of poly-α-glutamic acid and derivatives thereof
US8354549B2 (en) 2006-11-30 2013-01-15 Nektar Therapeutics Method for preparing a polymer conjugate
US8394365B2 (en) 2003-09-17 2013-03-12 Nektar Therapeutics Multi-arm polymer prodrugs
US8491880B2 (en) 2008-12-10 2013-07-23 Mersana Therapeutics, Inc. Pharmaceutical formulations of biodegradable biocompatible camptothecin-polymer conjugates
CN103263675A (en) * 2013-05-16 2013-08-28 湘潭大学 Poly (epsilon-caprolactone) supported anti-tumor prodrug and preparation method thereof
US8557961B2 (en) 2010-04-02 2013-10-15 Amunix Operating Inc. Alpha 1-antitrypsin compositions and methods of making and using same
US8563527B2 (en) 2007-08-20 2013-10-22 Pharmain Corporation Oligonucleotide core carrier compositions for delivery of nucleic acid-containing therapeutic agents, methods of making and using the same
US8574259B2 (en) 2005-05-10 2013-11-05 Lifescreen Sciences Llc Intravascular filter with drug reservoir
TWI417114B (en) * 2005-08-31 2013-12-01 Abraxis Bioscience Llc Compositions and methods for preparation of poorly water soluble drugs with increased stability
US8637466B2 (en) 2008-08-11 2014-01-28 Nektar Therapeutics Multi-arm polymeric alkanoate conjugates
US8673860B2 (en) 2009-02-03 2014-03-18 Amunix Operating Inc. Extended recombinant polypeptides and compositions comprising same
US8680050B2 (en) 2009-02-03 2014-03-25 Amunix Operating Inc. Growth hormone polypeptides fused to extended recombinant polypeptides and methods of making and using same
US8692002B2 (en) 2004-11-18 2014-04-08 Cardiome Pharma Corp. Synthetic process for aminocyclohexyl ether compounds
US8703717B2 (en) 2009-02-03 2014-04-22 Amunix Operating Inc. Growth hormone polypeptides and methods of making and using same
US8716448B2 (en) 2009-02-03 2014-05-06 Amunix Operating Inc. Coagulation factor VII compositions and methods of making and using same
US8840879B2 (en) 2004-03-11 2014-09-23 Fresenius Kabi Deutschland Gmbh Conjugates of hydroxyalkyl starch and a protein
US8889211B2 (en) 2010-09-02 2014-11-18 Boston Scientific Scimed, Inc. Coating process for drug delivery balloons using heat-induced rewrap memory
US8906353B2 (en) 2008-09-23 2014-12-09 Nektar Therapeutics Compositions and methods for achieving sustained therapeutic drug concentrations in a subject
US8916518B2 (en) 2002-03-06 2014-12-23 Fresenius Kabi Deutschland Gmbh Coupling proteins to a modified polysaccharide
US8933197B2 (en) 2007-08-15 2015-01-13 Amunix Operating Inc. Compositions comprising modified biologically active polypeptides
US8957021B2 (en) 2009-02-03 2015-02-17 Amunix Operating Inc. Glucose-regulating polypeptides and methods of making and using same
US8999930B2 (en) 2008-01-09 2015-04-07 Pharmain Corporation Soluble hydrophobic core carrier compositions for delivery of therapeutic agents, methods of making and using the same
US9050378B2 (en) 2003-12-10 2015-06-09 Board Of Regents, The University Of Texas System N2S2 chelate-targeting ligand conjugates
US9056152B2 (en) 2011-08-25 2015-06-16 Boston Scientific Scimed, Inc. Medical device with crystalline drug coating
US9192697B2 (en) 2007-07-03 2015-11-24 Hemoteq Ag Balloon catheter for treating stenosis of body passages and for preventing threatening restenosis
US9498488B2 (en) 2003-06-27 2016-11-22 Biorest Ltd. Method of treating acute coronary syndromes
US9550860B2 (en) 2002-09-06 2017-01-24 Cerulean Pharma Inc. Cyclodextrin-based polymers for therapeutics delivery
EP2068956B1 (en) * 2006-06-26 2017-03-01 Boston Scientific Limited Medical devices for release of low solubility therapeutic agents
US9610360B2 (en) 2007-01-24 2017-04-04 Ceruliean Pharma Inc. Polymer drug conjugates with tether groups for controlled drug delivery
US9649476B2 (en) 2002-09-20 2017-05-16 Bayer Intellectual Property Gmbh Medical device for dispersing medicaments
US9730820B2 (en) 2008-09-25 2017-08-15 Abbott Cardiovascular Systems Inc. Stent delivery system having a fibrous matrix covering with improved stent retention
US9737615B2 (en) 2005-12-19 2017-08-22 PharmalN Corporation Hydrophobic core carrier compositions for delivery of therapeutic agents, methods of making and using the same
US9849188B2 (en) 2009-06-08 2017-12-26 Amunix Operating Inc. Growth hormone polypeptides and methods of making and using same
US9855338B2 (en) 2005-12-05 2018-01-02 Nitto Denko Corporation Polyglutamate-amino acid conjugates and methods
US9901663B2 (en) 2013-05-06 2018-02-27 Abbott Cardiovascular Systems Inc. Hollow stent filled with a therapeutic agent formulation
US9993427B2 (en) 2013-03-14 2018-06-12 Biorest Ltd. Liposome formulation and manufacture
EP3372617A2 (en) 2010-04-02 2018-09-12 Amunix Operating Inc. Binding fusion proteins, binding fusion protein-drug conjugates, xten-drug conjugates and methods of making and using same
US10080821B2 (en) 2009-07-17 2018-09-25 Boston Scientific Scimed, Inc. Nucleation of drug delivery balloons to provide improved crystal size and density
US10098865B2 (en) 2010-12-22 2018-10-16 Nektar Therapeutics Multi-arm polymeric prodrug conjugates of taxane-based compounds
US10172953B2 (en) 2012-02-27 2019-01-08 Amunix Operating Inc. XTEN conjugate compositions and methods of making same
US10370430B2 (en) 2012-02-15 2019-08-06 Bioverativ Therapeutics Inc. Recombinant factor VIII proteins
US10369256B2 (en) 2009-07-10 2019-08-06 Boston Scientific Scimed, Inc. Use of nanocrystals for drug delivery from a balloon
US10421798B2 (en) 2012-02-15 2019-09-24 Bioverativ Therapeutics Inc. Factor VIII compositions and methods of making and using same
US10548953B2 (en) 2013-08-14 2020-02-04 Bioverativ Therapeutics Inc. Factor VIII-XTEN fusions and uses thereof
US10745680B2 (en) 2015-08-03 2020-08-18 Bioverativ Therapeutics Inc. Factor IX fusion proteins and methods of making and using same
US10814013B2 (en) 2006-10-05 2020-10-27 The Board Of Regents Of The University Of Texas System Efficient synthesis of chelators for nuclear imaging and radiotherapy: compositions and applications
US10894087B2 (en) 2010-12-22 2021-01-19 Nektar Therapeutics Multi-arm polymeric prodrug conjugates of cabazitaxel-based compounds
US11464871B2 (en) 2012-10-02 2022-10-11 Novartis Ag Methods and systems for polymer precipitation and generation of particles
EP3917397A4 (en) * 2019-01-28 2022-10-26 Board of Regents, The University of Texas System Metal chelator combination therapy for the treatment of cancer
US11713358B2 (en) 2015-08-28 2023-08-01 Amunix Pharmaceuticals, Inc. Chimeric polypeptide assembly and methods of making and using the same

Families Citing this family (256)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6096331A (en) * 1993-02-22 2000-08-01 Vivorx Pharmaceuticals, Inc. Methods and compositions useful for administration of chemotherapeutic agents
US20030133955A1 (en) * 1993-02-22 2003-07-17 American Bioscience, Inc. Methods and compositions useful for administration of chemotherapeutic agents
US6749868B1 (en) 1993-02-22 2004-06-15 American Bioscience, Inc. Protein stabilized pharmacologically active agents, methods for the preparation thereof and methods for the use thereof
US6753006B1 (en) 1993-02-22 2004-06-22 American Bioscience, Inc. Paclitaxel-containing formulations
US20030068362A1 (en) * 1993-02-22 2003-04-10 American Bioscience, Inc. Methods and formulations for the delivery of pharmacologically active agents
US6537579B1 (en) 1993-02-22 2003-03-25 American Bioscience, Inc. Compositions and methods for administration of pharmacologically active compounds
US6774278B1 (en) 1995-06-07 2004-08-10 Cook Incorporated Coated implantable medical device
DK0914102T3 (en) * 1996-05-24 2006-01-09 Angiotech Pharm Inc Preparations and methods for treating or preventing diseases of the body canals
US20070092563A1 (en) * 1996-10-01 2007-04-26 Abraxis Bioscience, Inc. Novel formulations of pharmacological agents, methods for the preparation thereof and methods for the use thereof
US6495579B1 (en) 1996-12-02 2002-12-17 Angiotech Pharmaceuticals, Inc. Method for treating multiple sclerosis
US20030157187A1 (en) * 1996-12-02 2003-08-21 Angiotech Pharmaceuticals, Inc. Compositions and methods for treating or preventing inflammatory diseases
US6515016B2 (en) 1996-12-02 2003-02-04 Angiotech Pharmaceuticals, Inc. Composition and methods of paclitaxel for treating psoriasis
US6204388B1 (en) 1996-12-03 2001-03-20 Sloan-Kettering Institute For Cancer Research Synthesis of epothilones, intermediates thereto and analogues thereof
CA2273083C (en) 1996-12-03 2012-09-18 Sloan-Kettering Institute For Cancer Research Synthesis of epothilones, intermediates thereto, analogues and uses thereof
US7112338B2 (en) * 1997-03-12 2006-09-26 The Regents Of The University Of California Cationic liposome delivery of taxanes to angiogenic blood vessels
DE19718339A1 (en) * 1997-04-30 1998-11-12 Schering Ag Polymer coated stents, processes for their manufacture and their use for restenosis prophylaxis
US20030199425A1 (en) * 1997-06-27 2003-10-23 Desai Neil P. Compositions and methods for treatment of hyperplasia
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
US20040170563A1 (en) * 1997-10-27 2004-09-02 Meade Thomas J. Magnetic resonance imaging agents for the delivery of therapeutic agents
US6485514B1 (en) * 1997-12-12 2002-11-26 Supergen, Inc. Local delivery of therapeutic agents
US6394945B1 (en) * 1997-12-22 2002-05-28 Mds (Canada), Inc. Radioactively coated devices
KR100228187B1 (en) * 1997-12-24 1999-11-01 김성년 A radioactive ballon used in balloon dilatation catherer and process for preparation thereof
US7314637B1 (en) 1999-06-29 2008-01-01 Neopharm, Inc. Method of administering liposomal encapsulated taxane
US6350786B1 (en) 1998-09-22 2002-02-26 Hoffmann-La Roche Inc. Stable complexes of poorly soluble compounds in ionic polymers
US7018654B2 (en) * 1999-03-05 2006-03-28 New River Pharmaceuticals Inc. Pharmaceutical composition containing an active agent in an amino acid copolymer structure
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
US20040121954A1 (en) * 1999-04-13 2004-06-24 Xu Wuhan Jingya Poly(dipeptide) as a drug carrier
US6317615B1 (en) 1999-04-19 2001-11-13 Cardiac Pacemakers, Inc. Method and system for reducing arterial restenosis in the presence of an intravascular stent
US6368658B1 (en) * 1999-04-19 2002-04-09 Scimed Life Systems, Inc. Coating medical devices using air suspension
DE60036382D1 (en) * 1999-06-21 2007-10-25 Nihon Mediphysics Co Ltd METHOD FOR THE ADMINISTRATION OF MEDICAMENTS WITH BINDING SAFFINITY TO PLASMA PROTEIN AND USE OF THE COMPOSITION IN THE PROCESS
US6258121B1 (en) * 1999-07-02 2001-07-10 Scimed Life Systems, Inc. Stent coating
EE200200127A (en) * 1999-09-09 2003-04-15 The Regents Of The University Of California Delivering taxanes to angiogenic blood vessels with cationic liposomes
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
CA2385528C (en) 1999-10-01 2013-12-10 Immunogen, Inc. Compositions and methods for treating cancer using immunoconjugates and chemotherapeutic agents
US20030054977A1 (en) * 1999-10-12 2003-03-20 Cell Therapeutics, Inc. Manufacture of polyglutamate-therapeutic agent conjugates
US6692724B1 (en) 1999-10-25 2004-02-17 Board Of Regents, The University Of Texas System Ethylenedicysteine (EC)-drug conjugates, compositions and methods for tissue specific disease imaging
US6362217B2 (en) * 2000-03-17 2002-03-26 Bristol-Myers Squibb Company Taxane anticancer agents
AU2001253479A1 (en) 2000-04-13 2001-10-30 Sts Biopolymers, Inc. Targeted therapeutic agent release devices and methods of making and using the same
US8236048B2 (en) 2000-05-12 2012-08-07 Cordis Corporation Drug/drug delivery systems for the prevention and treatment of vascular disease
US6776796B2 (en) 2000-05-12 2004-08-17 Cordis Corportation Antiinflammatory drug and delivery device
CN1292798C (en) * 2000-06-02 2007-01-03 德克萨斯州立大学董事会 Ethylenedicysteine (EC)-drug conjugates
CN1125097C (en) * 2000-07-05 2003-10-22 天津大学 Precursor of polyglycol carried taxusol or polyene taxusol
US20020099013A1 (en) * 2000-11-14 2002-07-25 Thomas Piccariello Active agent delivery systems and methods for protecting and administering active agents
US7163918B2 (en) 2000-08-22 2007-01-16 New River Pharmaceuticals Inc. Iodothyronine compositions
WO2005032474A2 (en) * 2003-09-30 2005-04-14 New River Pharmaceuticals Inc. Pharmaceutical compositions for prevention of overdose or abuse
WO2003079972A2 (en) 2002-02-22 2003-10-02 New River Parmaceuticals Inc. Active agent delivery systems and methods for protecting and administering active agents
US20060177416A1 (en) 2003-10-14 2006-08-10 Medivas, Llc Polymer particle delivery compositions and methods of use
ATE343969T1 (en) 2000-09-29 2006-11-15 Cordis Corp COATED MEDICAL DEVICES
US6612976B2 (en) * 2000-11-13 2003-09-02 Isotech, L.L.C. Radioactive medical devices and methods of making radioactive medical devices
US6616592B1 (en) * 2000-11-13 2003-09-09 Isotech, L.L.C. Radioactive medical devices for inhibiting a hyperplastic response and method of making radioactive medical devices
US8394813B2 (en) 2000-11-14 2013-03-12 Shire Llc Active agent delivery systems and methods for protecting and administering active agents
CA2428971A1 (en) * 2000-11-14 2003-05-01 New River Pharmaceuticals Inc. Conjugates of a therapeutic agent and a peptide carrier
US20090306228A1 (en) * 2000-11-14 2009-12-10 Shire Llc Active agent delivery systems and methods for protecting and administering active agents
EP1347794A2 (en) * 2000-11-27 2003-10-01 Medtronic, Inc. Stents and methods for preparing stents from wires having hydrogel coating layers thereon
WO2002049501A2 (en) * 2000-12-18 2002-06-27 Board Of Regents, University Of Texas System Local regional chemotherapy and radiotherapy using in situ hydrogel
ATE290882T1 (en) 2001-01-16 2005-04-15 Glaxo Group Ltd PHARMACEUTICAL COMPOUND AGAINST CANCER CONTAINING A 4-QUINAZOLINAMININE IN COMBINATION WITH PACLITAXEL, CARBOPLATIN OR VINORELBINE
KR20030071853A (en) * 2001-01-25 2003-09-06 브리스톨-마이어스스퀴브컴파니 Parenteral Formulations Containing Epothilone Analogs
WO2002058699A1 (en) * 2001-01-25 2002-08-01 Bristol-Myers Squibb Company Pharmaceutical forms of epothilones for oral administration
US7771468B2 (en) * 2001-03-16 2010-08-10 Angiotech Biocoatings Corp. Medicated stent having multi-layer polymer coating
PT1372650E (en) * 2001-03-19 2009-02-25 Novartis Ag Combinations comprising an antidiarrheal agent and an epothilone or an epothilone derivative
PE20020908A1 (en) * 2001-03-21 2002-10-26 Cell Therapeutics Inc RECOMBINANT PRODUCTION OF POLYANIONIC POLYMERS AND USE OF THE SAME
CA2444383A1 (en) * 2001-04-26 2002-11-07 Board Of Regents, The University Of Texas System Therapeutic agent/ligand conjugate compositions, their methods of synthesis and use
US20040234497A1 (en) * 2001-05-04 2004-11-25 Yi Luo Hyaluronic acid containing bioconjugates:targeted delivery of anti-cancer drugs to cancer cells
US7375082B2 (en) * 2002-02-22 2008-05-20 Shire Llc Abuse-resistant hydrocodone compounds
US7169752B2 (en) * 2003-09-30 2007-01-30 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
US7338939B2 (en) * 2003-09-30 2008-03-04 New River Pharmaceuticals Inc. 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
US7708712B2 (en) * 2001-09-04 2010-05-04 Broncus Technologies, Inc. Methods and devices for maintaining patency of surgically created channels in a body organ
WO2003035135A1 (en) * 2001-09-24 2003-05-01 Boston Scientific Limited Optimized dosing for drug coated stents
US7488313B2 (en) * 2001-11-29 2009-02-10 Boston Scientific Scimed, Inc. Mechanical apparatus and method for dilating and delivering a therapeutic agent to a site of treatment
DE10158904A1 (en) * 2001-11-30 2003-06-12 Roche Diagnostics Gmbh Process for the preparation of linear DNA fragments for the in vitro expression of proteins
KR20030049023A (en) * 2001-12-13 2003-06-25 주식회사 코오롱 Paclitaxel derivatives for radio-sensitizer
CN1273130C (en) * 2001-12-20 2006-09-06 布里斯托尔-迈尔斯斯奎布公司 Pharmaceutical compositions of orally active taxane derivatives having enhanced bioavailability
US7838619B2 (en) * 2002-01-14 2010-11-23 The General Hospital Corporation Biodegradable polyketal polymers and methods for their formation and use
KR20030068955A (en) * 2002-02-19 2003-08-25 주식회사 코오롱 New self-immolating linker and its preparation method, water-soluble prodrug compound comprising residue of paclitaxel or derivatives thereof using the same, its preparation method, and pharmaceutical composition comprising the same
US7105486B2 (en) * 2002-02-22 2006-09-12 New River Pharmaceuticals Inc. Abuse-resistant amphetamine compounds
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
ES2500117T3 (en) * 2002-02-22 2014-09-30 Shire Llc Novel sustained release pharmaceutical compounds to prevent the abuse of controlled substances
US7700561B2 (en) * 2002-02-22 2010-04-20 Shire Llc Abuse-resistant amphetamine prodrugs
US7659253B2 (en) 2002-02-22 2010-02-09 Shire Llc Abuse-resistant amphetamine prodrugs
CN100475269C (en) * 2002-03-05 2009-04-08 北京键凯科技有限公司 Binding agent of hydrophilic polymer-glutamic acid oligopeptide and medicinal molecular, composition containing said binding agent and use thereof
DE10209822A1 (en) * 2002-03-06 2003-09-25 Biotechnologie Ges Mittelhesse Coupling of low molecular weight substances to a modified polysaccharide
AU2003221291A1 (en) * 2002-03-13 2003-09-22 Beijing Jiankai Technology Co., Ltd. Hydrophilic polymer derivate with y type branch and preparation method of it medical composite comprising above compound
AU2003236076A1 (en) * 2002-03-22 2003-10-08 Beijing Jiankai Technology Co., Ltd. Hydrophilic polymers-flavonoids conjugates and pharmaceutical compositions comprising them
US7264822B2 (en) * 2002-04-03 2007-09-04 Poly-Med, Inc. Conjugated drug-polymer coated stent
JP2006514681A (en) * 2002-05-20 2006-05-11 コーザン バイオサイエンシス インコーポレイテッド Administration method of epothilone D
EP1521603B1 (en) 2002-07-12 2011-01-19 Cook Incorporated Coated medical device
US7649006B2 (en) 2002-08-23 2010-01-19 Sloan-Kettering Institute For Cancer Research Synthesis of epothilones, intermediates thereto and analogues thereof
DK1506203T3 (en) 2002-08-23 2007-05-14 Sloan Kettering Inst Cancer Synthesis of epothilones, intermediates thereof, analogs thereof and uses thereof
US20040047835A1 (en) * 2002-09-06 2004-03-11 Cell Therapeutics, Inc. Combinatorial drug therapy using polymer drug conjugates
IL166506A0 (en) * 2002-09-11 2006-01-15 Fresenius Kabi De Gmbh Hasylated polypeptides especially hasylated erythropoietin
WO2004037311A2 (en) * 2002-10-21 2004-05-06 Kensey Nash Corporation Device and methods for sequential, regional delivery of multiple cytotoxic agents
BR0316046A (en) * 2002-11-07 2005-09-13 Univ Texas Drug and ethylenodicysteine (ec) conjugates, compositions and methods for imaging tissue specific disease
AU2003291337B2 (en) 2002-11-07 2010-09-09 Kosan Biosciences Incorporated Trans-9,10-dehydroepothilone C and D, analogs thereof and methos of making the same
US20050004002A1 (en) 2002-12-09 2005-01-06 American Bioscience, Inc. Compositions and methods of delivery of pharmacological agents
WO2004063195A1 (en) * 2003-01-03 2004-07-29 Sloan-Kettering Institute For Cancer Research Pyridopyrimidine kinase inhibitors
CN1700906A (en) 2003-02-03 2005-11-23 新药物公司 Stable sterile filterable liposomal encapsulated taxane and other antineoplastic drugs
US7311727B2 (en) * 2003-02-05 2007-12-25 Board Of Trustees Of The University Of Arkansas Encased stent
KR101064901B1 (en) * 2003-03-20 2011-09-16 나노캐리어 가부시키가이샤 Micellar Preparation Containing Sparingly Water-soluble Anticancer Agent And Novel Block Copolymer
US7306580B2 (en) * 2003-04-16 2007-12-11 Cook Incorporated Medical device with therapeutic agents
KR100512483B1 (en) 2003-05-07 2005-09-05 선바이오(주) Novel Preparation method of PEG-maleimide PEG derivatives
TW200427503A (en) * 2003-05-27 2004-12-16 Kureha Chemical Ind Co Ltd Process for producing thermoplastic resin molding
EP1644019B2 (en) 2003-05-29 2018-02-21 Shire LLC Abuse resistant amphetamine compounds
US20050020557A1 (en) * 2003-05-30 2005-01-27 Kosan Biosciences, Inc. Method for treating diseases using HSP90-inhibiting agents in combination with enzyme inhibitors
US20050026893A1 (en) * 2003-05-30 2005-02-03 Kosan Biosciences, Inc. Method for treating diseases using HSP90-inhibiting agents in combination with immunosuppressants
US7691838B2 (en) 2003-05-30 2010-04-06 Kosan Biosciences Incorporated Method for treating diseases using HSP90-inhibiting agents in combination with antimitotics
US20050020534A1 (en) * 2003-05-30 2005-01-27 Kosan Biosciences, Inc. Method for treating diseases using HSP90-inhibiting agents in combination with antimetabolites
US20050054625A1 (en) * 2003-05-30 2005-03-10 Kosan Biosciences, Inc. Method for treating diseases using HSP90-inhibiting agents in combination with nuclear export inhibitors
US20050054589A1 (en) * 2003-05-30 2005-03-10 Kosan Biosciences, Inc. Method for treating diseases using HSP90-inhibiting agents in combination with antibiotics
US20050020556A1 (en) * 2003-05-30 2005-01-27 Kosan Biosciences, Inc. Method for treating diseases using HSP90-inhibiting agents in combination with platinum coordination complexes
SG145746A1 (en) * 2003-08-08 2008-09-29 Fresenius Kabi De Gmbh Conjugates of hydroxyalkyl starch and g-csf
US20050152979A1 (en) * 2003-09-05 2005-07-14 Cell Therapeutics, Inc. Hydrophobic drug compositions containing reconstitution enhancer
WO2005056636A2 (en) 2003-12-03 2005-06-23 Nektar Therapeutics Al, Corporation Method of preparing maleimide functionalized polymers
CN101659704A (en) 2004-03-11 2010-03-03 弗雷泽纽斯卡比德国有限公司 Conjugates of hydroxyalkyl starch and a protein, prepared by reductive amination
EP2279752A3 (en) * 2004-03-11 2011-08-24 Fresenius Kabi Deutschland GmbH Conjugates of hydroxyalkyl starch and a protein, prepared by native chemical ligation
GB0406445D0 (en) * 2004-03-23 2004-04-28 Astrazeneca Ab Combination therapy
KR20050099311A (en) * 2004-04-09 2005-10-13 에이엔에이치 케어연구소(주) Composition for injection comprising anticancer drug
WO2006004429A2 (en) * 2004-07-02 2006-01-12 Ge Healthcare As Imaging agents comprising a non- peptidic vector linked to a fluorophore via a polyethylene glycol linker
US8614228B2 (en) 2004-08-11 2013-12-24 Arqule, Inc. Quinone prodrug compositions and methods of use
EP1877097B1 (en) * 2004-08-11 2012-06-20 Arqule, Inc. Aminoacid conjugates of beta-lapachone for tumor targeting
RU2375384C2 (en) 2004-09-22 2009-12-10 Ниппон Каяку Кабусики Кайся New block copolymer, micellar preparation and anticarcinogenic agent containing micellar preparation as active component
US20120269886A1 (en) 2004-12-22 2012-10-25 Nitto Denko Corporation Therapeutic agent for pulmonary fibrosis
LT2727583T (en) 2004-12-22 2021-12-27 Nitto Denko Corporation Drug carrier and drug carrier kit for inhibiting fibrosis
DK2301531T3 (en) 2005-02-18 2018-07-30 Abraxis Bioscience Llc COMBINATIONS AND WAYS FOR ADMINISTRATING THERAPEUTIC SUBSTANCES AND COMBINATION THERAPY
CA2598528A1 (en) * 2005-03-11 2006-09-14 Fresenius Kabi Deutschland Gmbh Production of bioactive glycoproteins from inactive starting material
KR20080008364A (en) * 2005-05-05 2008-01-23 헤모텍 아게 All-over coating of vessel stents
WO2006122408A1 (en) 2005-05-18 2006-11-23 Aegera Therapeutics Inc. Bir domain binding compounds
JP5600240B2 (en) 2005-07-19 2014-10-01 ウェルズ ファーゴ バンク ナショナル アソシエイション Process for preparing polymeric maleimides
ITPD20050242A1 (en) 2005-08-03 2007-02-04 Fidia Farmaceutici BIOCONIUGATI ANTITUMORALI OF HYALURONIC ACID OR ITS DERIVATIVES, OBTAINABLE FOR DIRECT OR INDIRECT CHEMICAL CONJUGATION, AND THEIR USE IN PHARMACEUTICAL FIELD
EP1762250A1 (en) * 2005-09-12 2007-03-14 Fresenius Kabi Deutschland GmbH Conjugates of hydroxyalkyl starch and an active substance, prepared by chemical ligation via thiazolidine
WO2007038246A2 (en) 2005-09-22 2007-04-05 Medivas, Llc Solid polymer delivery compositions and methods for use thereof
JP5192384B2 (en) 2005-09-22 2013-05-08 メディバス エルエルシー Bis- (α-amino) -diol-diester-containing poly (ester amide) and poly (ester urethane) compositions and methods of use
CN100384419C (en) * 2005-12-02 2008-04-30 菏泽睿鹰制药集团有限公司 Epothilone slow-release implanting composition and use
US20070167349A1 (en) * 2005-12-06 2007-07-19 Cell Therapeutics, Inc. Estrogen cancer therapy
US9572886B2 (en) 2005-12-22 2017-02-21 Nitto Denko Corporation Agent for treating myelofibrosis
US8834912B2 (en) * 2005-12-30 2014-09-16 Boston Scientific Scimed, Inc. Medical devices having multiple charged layers
US7910152B2 (en) 2006-02-28 2011-03-22 Advanced Cardiovascular Systems, Inc. Poly(ester amide)-based drug delivery systems with controlled release rate and morphology
WO2007111211A1 (en) * 2006-03-28 2007-10-04 Nippon Kayaku Kabushiki Kaisha Polymer conjugate of taxane
US8758723B2 (en) 2006-04-19 2014-06-24 The Board Of Regents Of The University Of Texas System Compositions and methods for cellular imaging and therapy
SG10201407457UA (en) 2006-05-16 2014-12-30 Pharmascience Inc Iap bir domain binding compounds
RU2447095C2 (en) * 2006-05-18 2012-04-10 Ниппон Каяку Кабусики Кайся High-molecular weight conjugate of podophyllotoxins
MX2008016018A (en) 2006-06-15 2009-03-25 Cell Therapeutics Inc Sede Sec A process for the preparation of poly-alfa-glutamic acid and derivatives thereof.
EP1867657A1 (en) 2006-06-15 2007-12-19 Cell Therapeutics Europe S.R.L. Process for the preparation of poly-a-glutamic acid and derivatives thereof
AU2007269540B2 (en) * 2006-07-05 2013-06-27 Exelixis, Inc. Methods of using IGF1R and Abl kinase modulators
WO2008026048A2 (en) * 2006-08-31 2008-03-06 Wockhardt Research Centre Stable injectable pharmaceutical compositions of docetaxel
CA2664852A1 (en) * 2006-10-03 2008-04-10 Nippon Kayaku Kabushiki Kaisha High-molecular weight conjugate of resorcinol derivatives
US20080086195A1 (en) * 2006-10-05 2008-04-10 Boston Scientific Scimed, Inc. Polymer-Free Coatings For Medical Devices Formed By Plasma Electrolytic Deposition
WO2008056596A1 (en) * 2006-11-06 2008-05-15 Nippon Kayaku Kabushiki Kaisha Polymeric derivative of nucleic acid metabolic antagonist
EP2090607B1 (en) * 2006-11-08 2015-05-20 Nippon Kayaku Kabushiki Kaisha Polymeric derivative of nucleic acid metabolic antagonist
US8998846B2 (en) 2006-11-20 2015-04-07 Lutonix, Inc. Drug releasing coatings for balloon catheters
US9737640B2 (en) 2006-11-20 2017-08-22 Lutonix, Inc. Drug releasing coatings for medical devices
US20080276935A1 (en) 2006-11-20 2008-11-13 Lixiao Wang Treatment of asthma and chronic obstructive pulmonary disease with anti-proliferate and anti-inflammatory drugs
US8430055B2 (en) 2008-08-29 2013-04-30 Lutonix, Inc. Methods and apparatuses for coating balloon catheters
US8414910B2 (en) 2006-11-20 2013-04-09 Lutonix, Inc. Drug releasing coatings for medical devices
US8425459B2 (en) * 2006-11-20 2013-04-23 Lutonix, Inc. Medical device rapid drug releasing coatings comprising a therapeutic agent and a contrast agent
US8414525B2 (en) 2006-11-20 2013-04-09 Lutonix, Inc. Drug releasing coatings for medical devices
US8414526B2 (en) 2006-11-20 2013-04-09 Lutonix, Inc. Medical device rapid drug releasing coatings comprising oils, fatty acids, and/or lipids
US9700704B2 (en) 2006-11-20 2017-07-11 Lutonix, Inc. Drug releasing coatings for balloon catheters
US20080175887A1 (en) 2006-11-20 2008-07-24 Lixiao Wang Treatment of Asthma and Chronic Obstructive Pulmonary Disease With Anti-proliferate and Anti-inflammatory Drugs
CN101209350B (en) 2006-12-30 2011-09-07 中国人民解放军军事医学科学院毒物药物研究所 Polyglutamate-medicament coupling compound with amino acid as communicating terminal
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
US20080181852A1 (en) * 2007-01-29 2008-07-31 Nitto Denko Corporation Multi-functional Drug Carriers
US8784866B2 (en) 2007-03-26 2014-07-22 William Marsh Rice University Water-soluble carbon nanotube compositions for drug delivery and medicinal applications
TWI407971B (en) 2007-03-30 2013-09-11 Nitto Denko Corp Cancer cells and tumor-related fibroblasts
CN104800856A (en) * 2007-04-10 2015-07-29 日东电工株式会社 Multi-functional polyglutamate drug carriers
WO2008134528A1 (en) * 2007-04-25 2008-11-06 Board Of Regents, The University Of Texas System Anti-cancer agent-hyaluronic acid conjugate compositions and methods
EP2152686B1 (en) 2007-04-30 2014-12-17 ArQule, Inc. Hydroxy sulfonate of quinone compounds and their uses
US20080279778A1 (en) * 2007-05-09 2008-11-13 Nitto Denko Corporation Polyglutamate conjugates and polyglutamate-amino acid conjugates having a plurality of drugs
US20080279782A1 (en) * 2007-05-09 2008-11-13 Nitto Denko Corporation Polymers conjugated with platinum drugs
US8197828B2 (en) * 2007-05-09 2012-06-12 Nitto Denko Corporation Compositions that include a hydrophobic compound and a polyamino acid conjugate
US8252361B2 (en) * 2007-06-05 2012-08-28 Abbott Cardiovascular Systems Inc. Implantable medical devices for local and regional treatment
US10092524B2 (en) 2008-06-11 2018-10-09 Edge Therapeutics, Inc. Compositions and their use to treat complications of aneurysmal subarachnoid hemorrhage
NZ581836A (en) 2007-06-11 2012-07-27 R Loch Macdonald A drug delivery system for the prevention of cerebral vasospasm
JP2010539245A (en) * 2007-09-14 2010-12-16 日東電工株式会社 Drug carrier
WO2009041570A1 (en) * 2007-09-28 2009-04-02 Nippon Kayaku Kabushiki Kaisha Polymer conjugate of steroid
WO2009070380A2 (en) * 2007-10-03 2009-06-04 William Marsh Rice University Water-soluble carbon nanotube compositions for drug delivery and medical applications
EP2070951A1 (en) * 2007-12-14 2009-06-17 Fresenius Kabi Deutschland GmbH Method for producing a hydroxyalkyl starch derivatives with two linkers
US20090169480A1 (en) * 2007-12-31 2009-07-02 Industrial Technology Research Institute Dendritic polymers and magnetic resonance imaging contrast agent employing the same
US8101706B2 (en) 2008-01-11 2012-01-24 Serina Therapeutics, Inc. Multifunctional forms of polyoxazoline copolymers and drug compositions comprising the same
CN102083468A (en) * 2008-03-06 2011-06-01 日东电工株式会社 Polymer paclitaxel conjugates and methods for treating cancer
EP2258397B1 (en) * 2008-03-18 2017-10-11 Nippon Kayaku Kabushiki Kaisha Polymer conjugate of physiologically active substance
CN101569747B (en) * 2008-04-30 2012-08-22 宁波大学 Preparation method of taxol prodrug using polyethylene glycol as carrier
CN101569748B (en) * 2008-04-30 2012-08-22 宁波大学 Water-soluble taxol prodrug prodrug preparation method
EP3159033B1 (en) * 2008-05-01 2019-02-27 Bayer Intellectual Property GmbH Catheter balloon drug adherence techniques and methods
JP5366940B2 (en) 2008-05-08 2013-12-11 日本化薬株式会社 Polymer conjugate of folic acid or folic acid derivative
JP2011162569A (en) * 2008-05-23 2011-08-25 Nano Career Kk Camptothecin polymer derivative and use thereof
AU2008359989A1 (en) * 2008-07-30 2010-02-04 Nitto Denko Corporation Drug carriers
US8049061B2 (en) 2008-09-25 2011-11-01 Abbott Cardiovascular Systems, Inc. Expandable member formed of a fibrous matrix having hydrogel polymer for intraluminal drug delivery
US8076529B2 (en) 2008-09-26 2011-12-13 Abbott Cardiovascular Systems, Inc. Expandable member formed of a fibrous matrix for intraluminal drug delivery
US8226603B2 (en) 2008-09-25 2012-07-24 Abbott Cardiovascular Systems Inc. Expandable member having a covering formed of a fibrous matrix for intraluminal drug delivery
US9060931B2 (en) 2008-10-31 2015-06-23 The Invention Science Fund I, Llc Compositions and methods for delivery of frozen particle adhesives
US9072799B2 (en) 2008-10-31 2015-07-07 The Invention Science Fund I, Llc Compositions and methods for surface abrasion with frozen particles
US8788211B2 (en) 2008-10-31 2014-07-22 The Invention Science Fund I, Llc Method and system for comparing tissue ablation or abrasion data to data related to administration of a frozen particle composition
US20100111857A1 (en) 2008-10-31 2010-05-06 Boyden Edward S Compositions and methods for surface abrasion with frozen particles
US8798933B2 (en) 2008-10-31 2014-08-05 The Invention Science Fund I, Llc Frozen compositions and methods for piercing a substrate
US8545806B2 (en) 2008-10-31 2013-10-01 The Invention Science Fund I, Llc Compositions and methods for biological remodeling with frozen particle compositions
US9050070B2 (en) 2008-10-31 2015-06-09 The Invention Science Fund I, Llc Compositions and methods for surface abrasion with frozen particles
US9072688B2 (en) 2008-10-31 2015-07-07 The Invention Science Fund I, Llc Compositions and methods for therapeutic delivery with frozen particles
US8849441B2 (en) 2008-10-31 2014-09-30 The Invention Science Fund I, Llc Systems, devices, and methods for making or administering frozen particles
US9050317B2 (en) 2008-10-31 2015-06-09 The Invention Science Fund I, Llc Compositions and methods for therapeutic delivery with frozen particles
US8721583B2 (en) 2008-10-31 2014-05-13 The Invention Science Fund I, Llc Compositions and methods for surface abrasion with frozen particles
US8725420B2 (en) 2008-10-31 2014-05-13 The Invention Science Fund I, Llc Compositions and methods for surface abrasion with frozen particles
US8793075B2 (en) 2008-10-31 2014-07-29 The Invention Science Fund I, Llc Compositions and methods for therapeutic delivery with frozen particles
US9060934B2 (en) 2008-10-31 2015-06-23 The Invention Science Fund I, Llc Compositions and methods for surface abrasion with frozen particles
US20100111841A1 (en) * 2008-10-31 2010-05-06 Searete Llc Compositions and methods for surface abrasion with frozen particles
US8731841B2 (en) 2008-10-31 2014-05-20 The Invention Science Fund I, Llc Compositions and methods for therapeutic delivery with frozen particles
US8762067B2 (en) 2008-10-31 2014-06-24 The Invention Science Fund I, Llc Methods and systems for ablation or abrasion with frozen particles and comparing tissue surface ablation or abrasion data to clinical outcome data
US9050251B2 (en) 2008-10-31 2015-06-09 The Invention Science Fund I, Llc Compositions and methods for delivery of frozen particle adhesives
US8731840B2 (en) 2008-10-31 2014-05-20 The Invention Science Fund I, Llc Compositions and methods for therapeutic delivery with frozen particles
US9060926B2 (en) 2008-10-31 2015-06-23 The Invention Science Fund I, Llc Compositions and methods for therapeutic delivery with frozen particles
JP5544357B2 (en) 2009-05-15 2014-07-09 日本化薬株式会社 Polymer conjugate of a physiologically active substance having a hydroxyl group
CA2781669A1 (en) * 2009-11-23 2011-05-26 Cerulean Pharma Inc. Cyclodextrin-based polymers for therapeutic delivery
US20110160645A1 (en) * 2009-12-31 2011-06-30 Boston Scientific Scimed, Inc. Cryo Activated Drug Delivery and Cutting Balloons
CN102985439B9 (en) 2010-02-12 2016-08-03 制药科学股份有限公司 IAP BIR domain binding compounds
WO2011123395A1 (en) 2010-03-29 2011-10-06 Abraxis Bioscience, Llc Methods of treating cancer
CN107158389A (en) 2010-03-29 2017-09-15 阿布拉科斯生物科学有限公司 Strengthen the method for medicine delivery and therapeutic agent validity
DE102010022588A1 (en) 2010-05-27 2011-12-01 Hemoteq Ag Balloon catheter with a particle-free drug-releasing coating
EP2575804A4 (en) 2010-06-04 2013-10-23 Abraxis Bioscience Llc Methods of treatment of pancreatic cancer
US20140088298A9 (en) 2010-07-09 2014-03-27 Fresenius Kabi Deutschland Gmbh Conjugates comprising hydroxyalkyl starch and a cytotoxic agent and process for their preparation
US20130217871A1 (en) 2010-07-09 2013-08-22 Helmut Knoller Conjugates comprising hydroxyalkyl starch and a cytotoxic agent and process for their preparation
EP2591008A1 (en) 2010-07-09 2013-05-15 Fresenius Kabi Deutschland GmbH Conjugates comprising hydroxyalkyl starch and a cytotoxic agent and process for their preparation
EP2590680A1 (en) 2010-07-09 2013-05-15 Fresenius Kabi Deutschland GmbH Conjugates comprising hydroxyalkyl starch and a cytotoxic agent and process for their preparation
CN102339813A (en) 2010-07-14 2012-02-01 中国科学院微电子研究所 Semiconductor structure and manufacturing method thereof
CA2816997A1 (en) 2010-11-17 2012-05-24 Nippon Kayaku Kabushiki Kaisha Novel polymer derivative of cytidine metabolic antagonist
JP6146814B2 (en) 2011-02-11 2017-06-14 エッジ セラピュティックス インコーポレイテッド Compositions and methods for improving human prognosis of subarachnoid hemorrhage
KR101302698B1 (en) * 2011-02-28 2013-09-03 부산대학교 산학협력단 Drug-eluting stent using block copolymers composed of poly(hydroxyoctanoate) and biodegradable polymer
KR101302703B1 (en) 2011-02-28 2013-09-03 부산대학교 산학협력단 Drug eluting stent of graft copolymers composed of poly(3-hydroxyoctanoate) and biodegradable polymer
KR101328660B1 (en) * 2011-02-28 2013-11-14 부산대학교 산학협력단 Sorafenib-incorporated poly(ε-caprolactone) film and its drug-eluting stent
AU2012240131B2 (en) 2011-04-05 2017-07-20 Edge Therapeutics Intraventricular drug delivery system for improving outcome after a brain injury affecting cerebral blood flow
US9873765B2 (en) 2011-06-23 2018-01-23 Dsm Ip Assets, B.V. Biodegradable polyesteramide copolymers for drug delivery
CA2839526A1 (en) 2011-06-23 2012-12-27 Dsm Ip Assets B.V. Micro- or nanoparticles comprising a biodegradable polyesteramide copolymer for use in the delivery of bioactive agents
CN102850301A (en) * 2011-06-28 2013-01-02 中国人民解放军军事医学科学院毒物药物研究所 Water-soluble paclitaxel derivates and pharmaceutical composition and medicinal application thereof
WO2013022458A1 (en) 2011-08-05 2013-02-14 Boston Scientific Scimed, Inc. Methods of converting amorphous drug substance into crystalline form
US9346923B2 (en) 2011-09-11 2016-05-24 Nippon Kayaku Kabushiki Kaisha Method for manufacturing block copolymer
CN103083680B (en) 2011-11-07 2014-12-24 北京键凯科技有限公司 Polyethylene glycol (PEG)-amino acid oligopeptide-irinotecan combo and its medicinal composition
WO2013146381A1 (en) * 2012-03-27 2013-10-03 テルモ株式会社 Drug coat layer and medical instrument including same
KR101809858B1 (en) 2012-04-04 2017-12-15 할로자임, 아이엔씨 Combination therapy with an anti-hyaluronan agent and a tumor-targeted taxane
CN102614110B (en) * 2012-04-27 2013-12-25 北京大学 Stable polyethylene glycol medicinal micelle composition and preparation method thereof
US9399019B2 (en) 2012-05-09 2016-07-26 Evonik Corporation Polymorph compositions, methods of making, and uses thereof
CN102702140B (en) * 2012-06-19 2014-05-14 中国医学科学院生物医学工程研究所 Preparation method and application of water-soluble paclitaxel compound
CN102731442B (en) * 2012-07-18 2014-06-11 中国医学科学院生物医学工程研究所 Preparation method and application of water-soluble docetaxel compounds
AU2013352106B2 (en) 2012-11-30 2018-04-26 Novomedix, Llc Substituted biaryl sulfonamides and the use thereof
CN104721830A (en) * 2013-12-20 2015-06-24 北京蓝贝望生物医药科技股份有限公司 Top peptide
CA2969171C (en) 2014-12-18 2023-12-12 Dsm Ip Assets B.V. Drug delivery system for delivery of acid sensitive drugs
CN107683134B (en) * 2015-05-15 2021-02-23 珠海贝海生物技术有限公司 Docetaxel and human serum albumin complex
KR101726728B1 (en) * 2015-07-28 2017-04-14 주식회사 삼양바이오팜 Method for analyzing related substances of a pharmaceutical composition containing a polymeric carrier
CN106554330B (en) * 2015-09-26 2019-07-05 南京友怡医药科技有限公司 Water-soluble docetaxel anti-cancer drug compounds and its preparation method and application
CN106554329B (en) * 2015-09-26 2019-07-05 南京友怡医药科技有限公司 Water-soluble paclitaxel anti-cancer drug compounds and its preparation method and application
WO2017193757A1 (en) * 2016-05-10 2017-11-16 浙江海正药业股份有限公司 Water-soluble epothilone derivative and preparation method therefor
CN109415378B (en) * 2016-05-10 2021-11-09 浙江海正药业股份有限公司 Water-soluble Epothilone derivative and preparation method thereof
CN108478804B (en) * 2018-05-08 2020-09-22 辽宁大学 Polyacrylic acid-S-S-drug copolymer and preparation method thereof
WO2019222435A1 (en) 2018-05-16 2019-11-21 Halozyme, Inc. Methods of selecting subjects for combination cancer therapy with a polymer-conjugated soluble ph20
CN112604002A (en) * 2020-07-12 2021-04-06 苏州裕泰医药科技有限公司 Disulfide-bond bridged docetaxel-fatty acid prodrug and self-assembled nanoparticles thereof
US20230277490A1 (en) * 2020-07-31 2023-09-07 Cedars-Sinai Medical Center Glutamine as an anticancer therapy in solid tumors

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5169933A (en) * 1988-08-15 1992-12-08 Neorx Corporation Covalently-linked complexes and methods for enhanced cytotoxicity and imaging
US5362831A (en) * 1992-06-19 1994-11-08 Farmitalia Carlo Erba S.R.L. Polymer-bound paclitaxel derivatives
US5489525A (en) * 1992-10-08 1996-02-06 The United States Of America As Represented By The Department Of Health And Human Services Monoclonal antibodies to prostate cells
US5626862A (en) * 1994-08-02 1997-05-06 Massachusetts Institute Of Technology Controlled local delivery of chemotherapeutic agents for treating solid tumors

Family Cites Families (36)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4356166A (en) * 1978-12-08 1982-10-26 University Of Utah Time-release chemical delivery system
IN165717B (en) * 1986-08-07 1989-12-23 Battelle Memorial Institute
US4942184A (en) 1988-03-07 1990-07-17 The United States Of America As Represented By The Department Of Health And Human Services Water soluble, antineoplastic derivatives of taxol
US4960790A (en) * 1989-03-09 1990-10-02 University Of Kansas Derivatives of taxol, pharmaceutical compositions thereof and methods for the preparation thereof
US5219564A (en) * 1990-07-06 1993-06-15 Enzon, Inc. Poly(alkylene oxide) amino acid copolymers and drug carriers and charged copolymers based thereon
US5059699A (en) 1990-08-28 1991-10-22 Virginia Tech Intellectual Properties, Inc. Water soluble derivatives of taxol
US6515009B1 (en) * 1991-09-27 2003-02-04 Neorx Corporation Therapeutic inhibitor of vascular smooth muscle cells
US5811447A (en) 1993-01-28 1998-09-22 Neorx Corporation Therapeutic inhibitor of vascular smooth muscle cells
US5272171A (en) 1992-02-13 1993-12-21 Bristol-Myers Squibb Company Phosphonooxy and carbonate derivatives of taxol
JPH05286868A (en) 1992-04-03 1993-11-02 Kiyoshi Okawa Carcinostatic agent complex and its screening method
JPH069600A (en) 1992-05-06 1994-01-18 Bristol Myers Squibb Co Benzoate derivative of taxole
WO1993024476A1 (en) * 1992-06-04 1993-12-09 Clover Consolidated, Limited Water-soluble polymeric carriers for drug delivery
CA2086874E (en) 1992-08-03 2000-01-04 Renzo Mauro Canetta Methods for administration of taxol
US5614549A (en) * 1992-08-21 1997-03-25 Enzon, Inc. High molecular weight polymer-based prodrugs
WO1994005282A1 (en) 1992-09-04 1994-03-17 The Scripps Research Institute Water soluble taxol derivatives
US5380751A (en) 1992-12-04 1995-01-10 Bristol-Myers Squibb Company 6,7-modified paclitaxels
NO310238B1 (en) 1992-12-24 2001-06-11 Bristol Myers Squibb Co Chemical compounds and pharmaceutical preparations and the use of such preparations
US5981568A (en) 1993-01-28 1999-11-09 Neorx Corporation Therapeutic inhibitor of vascular smooth muscle cells
CA2154667A1 (en) * 1993-02-02 1994-08-18 Linda M. Gustavson Directed biodistribution of small molecules
AU6400094A (en) * 1993-03-09 1994-09-26 Enzon, Inc. Taxol polyalkylene oxide conjugates of taxol and taxol intermediates
WO1994020089A1 (en) * 1993-03-09 1994-09-15 Enzon, Inc. Taxol-based compositions with enhanced bioactivity
US5468769A (en) 1993-07-15 1995-11-21 Abbott Laboratories Paclitaxel derivatives
US5886026A (en) 1993-07-19 1999-03-23 Angiotech Pharmaceuticals Inc. Anti-angiogenic compositions and methods of use
WO1995003036A1 (en) * 1993-07-19 1995-02-02 Angiogenesis Technologies, Inc. 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
US5880131A (en) * 1993-10-20 1999-03-09 Enzon, Inc. High molecular weight polymer-based prodrugs
US5840900A (en) * 1993-10-20 1998-11-24 Enzon, Inc. High molecular weight polymer-based prodrugs
US5643575A (en) * 1993-10-27 1997-07-01 Enzon, Inc. Non-antigenic branched polymer conjugates
US5415869A (en) * 1993-11-12 1995-05-16 The Research Foundation Of State University Of New York Taxol formulation
US5730968A (en) * 1994-03-31 1998-03-24 Sterling Winthrop Inc. Segmented chelating polymers as imaging and therapeutic agents
US5583153A (en) * 1994-10-06 1996-12-10 Regents Of The University Of California Use of taxol in the treatment of rheumatoid arthritis
US5489589A (en) 1994-12-07 1996-02-06 Bristol-Myers Squibb Company Amino acid derivatives of paclitaxel
AU716005B2 (en) 1995-06-07 2000-02-17 Cook Medical Technologies Llc Implantable medical device
US5762909A (en) 1995-08-31 1998-06-09 General Electric Company Tumor targeting with polymeric molecules having extended conformation
NZ332234A (en) * 1996-03-12 2000-06-23 Pg Txl Company Lp Water soluble paclitaxel prodrugs formed by conjugating paclitaxel or docetaxel with a polyglutamic acid polymer and use for treating cancer
US5854382A (en) 1997-08-18 1998-12-29 Meadox Medicals, Inc. Bioresorbable compositions for implantable prostheses

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5169933A (en) * 1988-08-15 1992-12-08 Neorx Corporation Covalently-linked complexes and methods for enhanced cytotoxicity and imaging
US5362831A (en) * 1992-06-19 1994-11-08 Farmitalia Carlo Erba S.R.L. Polymer-bound paclitaxel derivatives
US5489525A (en) * 1992-10-08 1996-02-06 The United States Of America As Represented By The Department Of Health And Human Services Monoclonal antibodies to prostate cells
US5626862A (en) * 1994-08-02 1997-05-06 Massachusetts Institute Of Technology Controlled local delivery of chemotherapeutic agents for treating solid tumors

Cited By (241)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6179817B1 (en) 1995-02-22 2001-01-30 Boston Scientific Corporation Hybrid coating for medical devices
US7060724B2 (en) 1996-03-12 2006-06-13 Pg-Txl Company, L.P. Water soluble paclitaxel derivatives
US5977163A (en) * 1996-03-12 1999-11-02 Pg-Txl Company, L. P. Water soluble paclitaxel prodrugs
US6884817B2 (en) 1996-03-12 2005-04-26 Pg-Txl Company, L.P. Water soluble paclitaxel derivatives
US7384977B2 (en) 1996-03-12 2008-06-10 Pg-Txl Company, L.P. Water soluble paclitaxel prodrugs
US6441025B2 (en) 1996-03-12 2002-08-27 Pg-Txl Company, L.P. Water soluble paclitaxel derivatives
US7135496B2 (en) 1996-03-12 2006-11-14 Pg-Txl Company, L.P. Water soluble paclitaxel derivatives
EP0895784A4 (en) * 1996-04-15 2003-03-19 Asahi Chemical Ind Medicament composite
EP0895784A1 (en) * 1996-04-15 1999-02-10 Asahi Kasei Kogyo Kabushiki Kaisha Medicament composite
WO1997040854A2 (en) * 1996-05-01 1997-11-06 Antivirals Inc. Polypeptide conjugates for transporting substances across cell membranes
US6030941A (en) * 1996-05-01 2000-02-29 Avi Biopharma, Inc. Polymer composition for delivering substances in living organisms
WO1997040854A3 (en) * 1996-05-01 1998-03-12 Antivirals Inc Polypeptide conjugates for transporting substances across cell membranes
US6667048B1 (en) 1997-01-07 2003-12-23 Sonus Pharmaceuticals, Inc. Emulsion vehicle for poorly soluble drugs
US6660286B1 (en) 1997-01-07 2003-12-09 Sonus Pharmaceuticals, Inc. Emulsion vehicle for poorly soluble drugs
US6458373B1 (en) 1997-01-07 2002-10-01 Sonus Pharmaceuticals, Inc. Emulsion vehicle for poorly soluble drugs
US6982282B2 (en) 1997-01-07 2006-01-03 Sonus Pharmaceuticals, Inc. Emulsion vehicle for poorly soluble drugs
JP2002511845A (en) * 1997-04-24 2002-04-16 ニユコメド・イメージング・アクシエセルカペト Contrast agent
JP4993419B2 (en) * 1997-04-24 2012-08-08 ジーイー・ヘルスケア・アクスイェ・セルスカプ Contrast agent
US6495663B1 (en) 1997-05-21 2002-12-17 The Board Of Trustees Of The Leland Stanford Junior University Method and composition for enhancing transport across biological membranes
US6306993B1 (en) 1997-05-21 2001-10-23 The Board Of Trustees Of The Leland Stanford, Jr. University Method and composition for enhancing transport across biological membranes
WO1999016416A3 (en) * 1997-09-29 1999-08-05 Schering Ag Coated medical implants, method for producing them, and use of the same for preventing restenosis
WO1999016416A2 (en) * 1997-09-29 1999-04-08 Schering Ag Coated medical implants, method for producing them, and use of the same for preventing restenosis
JP2001526224A (en) * 1997-12-22 2001-12-18 シェーリング コーポレイション Combinations of benzocycloheptapyridine compounds and antitumor agents for treating proliferative diseases
US6555529B1 (en) 1997-12-25 2003-04-29 Toray Industries, Inc. Remedies for intramedullary diseases
WO1999033473A1 (en) * 1997-12-25 1999-07-08 Toray Industries, Inc. Remedies for intramedullary diseases
EP1800664A1 (en) * 1998-02-05 2007-06-27 Novartis AG Pharmaceutical formulations for parenteral administration comprising epothilone
BE1012140A3 (en) * 1998-02-05 2000-05-02 Novartis Ag Organic compounds.
WO1999039694A3 (en) * 1998-02-05 1999-09-23 Novartis Ag Compositions containing organic compounds
FR2774909A1 (en) * 1998-02-05 1999-08-20 Novartis Ag PHARMACEUTICAL FORMULATION, INFUSION SOLUTION AND LYOPHILIZED COMPOSITION COMPRISING EPOTHILONE.
WO1999039694A2 (en) * 1998-02-05 1999-08-12 Novartis Ag Compositions containing organic compounds
CN100396276C (en) * 1998-02-05 2008-06-25 诺瓦提斯公司 Compositions containing organic compounds
EP1028756A4 (en) * 1998-03-30 2002-05-08 Pg Txl Co Lp Water soluble paclitaxel derivatives
EP1598081A2 (en) * 1998-03-30 2005-11-23 PG-TXL Company, L.P. Conjugates of a water-soluble amino acid polymer and a drug
EP1028756A1 (en) * 1998-03-30 2000-08-23 PG-TXL Company, L.P. Water soluble paclitaxel derivatives
EP1598081A3 (en) * 1998-03-30 2008-05-07 PG-TXL Company, L.P. Conjugates of a water-soluble amino acid polymer and a drug
US7030155B2 (en) 1998-06-05 2006-04-18 Sonus Pharmaceuticals, Inc. Emulsion vehicle for poorly soluble drugs
JP2002519335A (en) * 1998-06-26 2002-07-02 クアナム メディカル コーポレイション Topoisomerase inhibitors for prevention of restenosis
WO2000000238A1 (en) * 1998-06-26 2000-01-06 Quanam Medical Corporation Topoisomerase inhibitors for prevention of restenosis
WO2000001682A1 (en) * 1998-07-01 2000-01-13 Bcm Developpement Inc. Water-soluble derivatives of paclitaxel, method for producing same and uses thereof
US6984400B2 (en) 1998-07-14 2006-01-10 Yissum Research Development Company Of The Hebrew University Of Jerusalem Method of treating restenosis using bisphosphonate nanoparticles
US7008645B2 (en) 1998-07-14 2006-03-07 Yissum Research Development Company Of The Hebrew University Of Jerusalem Method of inhibiting restenosis using bisphosphonates
EP1468691A1 (en) * 1998-07-14 2004-10-20 Yissum Research Development Company Of The Hebrew University Of Jerusalem Use of bisphosphonates in the treatment of vascular restenosis
WO2000003677A3 (en) * 1998-07-14 2000-05-04 Yissum Res Dev Co Use of bisphosphonates in the treatment of vascular restenosis
WO2000003677A2 (en) * 1998-07-14 2000-01-27 Yissum Research Development Company Of The Hebrew University Of Jerusalem Use of bisphosphonates in the treatment of vascular restenosis
AU758882B2 (en) * 1998-07-14 2003-04-03 Hadasit Medical Research Services & Development Company Ltd Treatment of restenosis
US6730064B2 (en) 1998-08-20 2004-05-04 Cook Incorporated Coated implantable medical device
US6299604B1 (en) 1998-08-20 2001-10-09 Cook Incorporated Coated implantable medical device
WO2000010622A1 (en) * 1998-08-20 2000-03-02 Cook Incorporated Coated implantable medical device
WO2000018439A3 (en) * 1998-09-29 2000-09-14 Schering Ag Use of neoangiogenesis markers for diagnosing and treating tumours by therapy
WO2000018439A2 (en) * 1998-09-29 2000-04-06 Schering Aktiengesellschaft Use of neoangiogenesis markers for diagnosing and treating tumours by therapy
EP1800702A2 (en) * 1998-10-14 2007-06-27 Boston Scientific Limited Loading and release of water-insoluble drugs
JP2002534461A (en) * 1999-01-12 2002-10-15 クアナム メディカル コーポレイション Compositions and methods for administration of water-insoluble paclitaxel derivatives
US6683100B2 (en) 1999-01-19 2004-01-27 Novartis Ag Organic compounds
WO2000044443A2 (en) * 1999-01-29 2000-08-03 Angiotech Pharmaceuticals Inc. Intra-pericardial delivery of anti-microtubule agents
WO2000044443A3 (en) * 1999-01-29 2001-08-16 Angiotech Pharm Inc Intra-pericardial delivery of anti-microtubule agents
US6333347B1 (en) 1999-01-29 2001-12-25 Angiotech Pharmaceuticals & Advanced Research Tech Intrapericardial delivery of anti-microtubule agents
WO2000050016A2 (en) * 1999-02-23 2000-08-31 Angiotech Pharmaceuticals, Inc. Compositions and methods for improving integrity of compromised body passageways and cavities
WO2000050016A3 (en) * 1999-02-23 2001-01-18 Angiotech Pharm Inc Compositions and methods for improving integrity of compromised body passageways and cavities
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
US6589266B2 (en) 1999-08-10 2003-07-08 Scimed Life Systems, Inc. Thrombosis filter having a surface treatment
US6273901B1 (en) 1999-08-10 2001-08-14 Scimed Life Systems, Inc. Thrombosis filter having a surface treatment
WO2001010342A1 (en) * 1999-08-10 2001-02-15 Scimed Life Systems, Inc. Thrombosis filter having a surface treatment
US8729010B2 (en) 1999-08-24 2014-05-20 Kai Pharmaceuticals, Inc. Compositions and methods for enhancing drug delivery across and into ocular tissues
US8278264B2 (en) 1999-08-24 2012-10-02 Kai Pharmaceuticals, Inc. Compositions and methods for enhancing drug delivery across and into ocular tissues
US8623833B2 (en) 1999-08-24 2014-01-07 Kai Pharmaceuticals, Inc. Compositions and methods for enhancing drug delivery across and into epithelial tissues
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
US6593292B1 (en) 1999-08-24 2003-07-15 Cellgate, Inc. Compositions and methods for enhancing drug delivery across and into epithelial tissues
US6759387B2 (en) 1999-08-24 2004-07-06 Cellgate, Inc. Compositions and methods for enhancing drug delivery across and into epithelial tissues
US6669951B2 (en) 1999-08-24 2003-12-30 Cellgate, Inc. Compositions and methods for enhancing drug delivery across and into epithelial tissues
US6541508B2 (en) 1999-09-13 2003-04-01 Nobex Corporation Taxane prodrugs
WO2001019406A2 (en) * 1999-09-13 2001-03-22 Nobex Corporation Amphiphilic prodrugs
US6380405B1 (en) 1999-09-13 2002-04-30 Nobex Corporation Taxane prodrugs
US7119074B2 (en) 1999-09-13 2006-10-10 Nobex Corporation Treatment of cancers, tumors and malignancies using amphiphilic prodrugs
WO2001019406A3 (en) * 1999-09-13 2002-01-17 Nobex Corp Amphiphilic prodrugs
US6713454B1 (en) 1999-09-13 2004-03-30 Nobex Corporation Prodrugs of etoposide and etoposide analogs
WO2001026693A3 (en) * 1999-10-12 2001-12-27 Cell Therapeutics Inc Manufacture of polyglutamate-therapeutic agent conjugates
SG147286A1 (en) * 1999-10-12 2008-11-28 Cell Therapeutics Inc Manufacture of polyglumate-therapeutic agent conjugates
WO2001026693A2 (en) * 1999-10-12 2001-04-19 Cell Therapeutics, Inc. Manufacture of polyglutamate-therapeutic agent conjugates
JP2003511423A (en) * 1999-10-12 2003-03-25 セル・セラピューティックス・インコーポレーテッド Preparation of polyglutamate-therapeutic drug conjugate
AU781735B2 (en) * 1999-10-12 2005-06-09 Cell Therapeutics, Inc. Manufacture of polyglutamate-therapeutic agent conjugates
US7399860B2 (en) 1999-10-12 2008-07-15 Cell Therapeutics Inc. Manufacture of polyglutamate-therapeutic agent conjugates
EP1466627A1 (en) * 1999-10-12 2004-10-13 Cell Therapeutics, Inc. Polyglutamate camptothecin conjugates
US7067111B1 (en) 1999-10-25 2006-06-27 Board Of Regents, University Of Texas System Ethylenedicysteine (EC)-drug conjugates, compositions and methods for tissue specific disease imaging
US7223380B2 (en) 1999-10-25 2007-05-29 Board Of Regents, The University Of Texas System Ethylenedicysteine (EC)-drug conjugates, compositions and methods for tissue specific disease imaging
US7632484B2 (en) 1999-10-25 2009-12-15 Board Of Regents, The University Of Texas System Ethylenedicysteine (EC)-drug conjugates, compositions and methods for tissue specific disease imaging
US7582281B2 (en) 1999-10-25 2009-09-01 Board Of Regents, The University Of Texas System Ethylenedicysteine (EC)-drug conjugates compositions and methods for tissue specific disease imaging
US7229604B2 (en) 1999-10-25 2007-06-12 Board Of Regents, The University Of Texas System Ethylenedicysteine (EC)-drug conjugates, compositions and methods for tissue specific disease imaging
US6638906B1 (en) 1999-12-13 2003-10-28 Nobex Corporation Amphiphilic polymers and polypeptide conjugates comprising same
JP2007153905A (en) * 1999-12-16 2007-06-21 F Hoffmann La Roche Ag Substituted bisindolylmaleimide for inhibition of cell proliferation
WO2001070275A3 (en) * 2000-03-17 2002-01-03 Cell Therapeutics Inc Polyglutamic acid-camptothecin conjugates and methods of preparation
US7153864B2 (en) 2000-03-17 2006-12-26 Cell Therapeutics Inc. Polyglutamic acid-camptothecin conjugates and methods of preparation
US7173041B2 (en) 2000-03-17 2007-02-06 Cell Therapeutics Inc. Polyglutamic acid-camptothecin conjugates and methods of preparation
WO2001070275A2 (en) * 2000-03-17 2001-09-27 Cell Therapeutics, Inc. Polyglutamic acid-camptothecin conjugates and methods of preparation
EP1301500A1 (en) * 2000-06-22 2003-04-16 Nitromed, Inc. Nitrosated and nitrosylated taxanes, compositions and methods of use
US6869973B2 (en) 2000-06-22 2005-03-22 Nitromed, Inc. Nitrosated and nitrosylated taxanes, compositions and methods of use
US6656966B2 (en) 2000-06-22 2003-12-02 Nitromed, Inc. Nitrosated and nitrosylated taxanes, compositions and methods of use
EP1301500A4 (en) * 2000-06-22 2004-03-31 Nitromed Inc Nitrosated and nitrosylated taxanes, compositions and methods of use
WO2002058688A1 (en) * 2001-01-24 2002-08-01 Mestex Ag Use of neurotoxic substances in producing a medicament for treating joint pains
AU2006222697B2 (en) * 2001-03-26 2008-03-13 Bayer Intellectual Property Gmbh Preparation for the Prophylaxis of Restenosis
US9066990B2 (en) 2001-03-26 2015-06-30 Bayer Intellectual Property Gmbh Preparation for restenosis prevention
AU2006222699B2 (en) * 2001-03-26 2008-04-03 Bayer Intellectual Property Gmbh Preparation for the Prophylaxis of Restenosis
WO2003037385A1 (en) * 2001-10-30 2003-05-08 Nektar Therapeutics Al, Corporation Water-soluble polymer conjugates of retinoic acid
US7261875B2 (en) 2001-12-21 2007-08-28 Board Of Regents, The University Of Texas System Dendritic poly (amino acid) carriers and methods of use
US8277776B2 (en) 2002-02-27 2012-10-02 Pharmain Corporation Compositions for delivery of therapeutics and other materials
US7138105B2 (en) 2002-02-27 2006-11-21 Pharmain Compositions for delivery of therapeutics and other materials, and methods of making and using the same
US8257682B2 (en) 2002-02-27 2012-09-04 Pharmain Corporation Compositions for delivery of therapeutics and other materials
EP1478406A1 (en) * 2002-02-27 2004-11-24 Pharmain, Ltd. Compositions for delivery of therapeutics and other materials, and methods of making and using the same
US8231859B2 (en) 2002-02-27 2012-07-31 Pharmain Corporation Compositions for delivery of therapeutics and other materials
EP1478406A4 (en) * 2002-02-27 2006-01-11 Pharmain Ltd Compositions for delivery of therapeutics and other materials, and methods of making and using the same
US8916518B2 (en) 2002-03-06 2014-12-23 Fresenius Kabi Deutschland Gmbh Coupling proteins to a modified polysaccharide
US9550860B2 (en) 2002-09-06 2017-01-24 Cerulean Pharma Inc. Cyclodextrin-based polymers for therapeutics delivery
US9649476B2 (en) 2002-09-20 2017-05-16 Bayer Intellectual Property Gmbh Medical device for dispersing medicaments
WO2004035629A3 (en) * 2002-10-18 2004-06-24 Fidia Farmaceutici Taxanes covalently bounded to hyaluronic acid or hyaluronic acid derivatives
WO2004035629A2 (en) * 2002-10-18 2004-04-29 Fidia Farmaceutici S.P.A. Taxanes covalently bounded to hyaluronic acid or hyaluronic acid derivatives
EP2045270A3 (en) * 2002-10-18 2009-06-03 FIDIA FARMACEUTICI S.p.A. Taxanes covalently bounded to hyaluronic acid or hyaluronic acid derivatives
HRP20050416B1 (en) * 2002-10-18 2013-09-30 Fidia Farmaceutici S.P.A. Taxanes covalently bounded to hyaluronic acid or hyaluronic acid derivatives
US7897584B2 (en) 2002-10-18 2011-03-01 Fidia Farmaceutici S.P.A. Taxanes covalently bounded to hyaluronic acid or hyaluronic acid derivatives
WO2004098651A3 (en) * 2003-05-09 2005-04-07 Schering Ag Bone localising radiopharmaceutical and tubulin-interacting compound combinatorial radiotherapy
EP1475105A1 (en) * 2003-05-09 2004-11-10 Schering AG Bone localising radiopharmaceutical and tubulin-interacting compound combinatorial radiotherapy
US7932377B2 (en) 2003-05-30 2011-04-26 Supramol Parenteral Colloids Gmbh Complexing of medicinal substances with high-molecular carriers and injection and infusion solutions containing said complexes
US10213446B2 (en) 2003-06-27 2019-02-26 Biorest Ltd. Method of treating acute coronary syndromes
US9498488B2 (en) 2003-06-27 2016-11-22 Biorest Ltd. Method of treating acute coronary syndromes
US10517883B2 (en) 2003-06-27 2019-12-31 Zuli Holdings Ltd. Method of treating acute myocardial infarction
US9827254B2 (en) 2003-06-27 2017-11-28 Biorest Ltd. Method of treating acute coronary syndromes
US8247427B2 (en) 2003-09-05 2012-08-21 The General Hospital Corporation Dual phase drug release system
US8101164B2 (en) 2003-09-05 2012-01-24 The General Hospital Corporation Dual phase drug release system
US8546419B2 (en) 2003-09-05 2013-10-01 The General Hospital Corporation Dual phase drug release system
US8394365B2 (en) 2003-09-17 2013-03-12 Nektar Therapeutics Multi-arm polymer prodrugs
US8318145B2 (en) 2003-09-17 2012-11-27 Nektar Therapeutics Multi-arm polymer prodrugs
US9333200B2 (en) 2003-09-17 2016-05-10 Nektar Therapeutics Multi-arm polymer prodrugs
US7744861B2 (en) 2003-09-17 2010-06-29 Nektar Therapeutics Multi-arm polymer prodrugs
US10463659B2 (en) 2003-09-17 2019-11-05 Nektar Therapeutics Multi-arm polymer prodrugs
US8771662B2 (en) 2003-09-17 2014-07-08 Nektar Therapeutics Multi-arm polymer prodrugs
US9808533B2 (en) 2003-09-17 2017-11-07 Nektar Therapeutics Multi-arm polymer prodrugs
US9050378B2 (en) 2003-12-10 2015-06-09 Board Of Regents, The University Of Texas System N2S2 chelate-targeting ligand conjugates
EP1561436A1 (en) * 2004-02-05 2005-08-10 SORIN BIOMEDICA CARDIO S.r.l. A stent for endoluminal delivery of active principles or agents
US8840879B2 (en) 2004-03-11 2014-09-23 Fresenius Kabi Deutschland Gmbh Conjugates of hydroxyalkyl starch and a protein
US7705036B2 (en) 2004-04-01 2010-04-27 Cardiome Pharma Corp. Deuterated aminocyclohexyl ether compounds and processes for preparing same
WO2005094897A2 (en) * 2004-04-01 2005-10-13 Cardiome Pharma Corp. Pegylated ion channel modulating compounds
US8022098B2 (en) 2004-04-01 2011-09-20 Cardiome Pharma Corp. Deuterated aminocyclohexyl ether compounds and processes for preparing same
WO2005094897A3 (en) * 2004-04-01 2006-06-22 Cardiome Pharma Corp Pegylated ion channel modulating compounds
US7786119B2 (en) 2004-04-01 2010-08-31 Cardiome Pharma Corp. Drug conjugates of ion channel modulating compounds
US8692002B2 (en) 2004-11-18 2014-04-08 Cardiome Pharma Corp. Synthetic process for aminocyclohexyl ether compounds
US9586899B2 (en) 2004-11-18 2017-03-07 Cardiome Pharma Corp. Synthetic process for aminocyclohexyl ether compounds
US9115081B2 (en) 2004-11-18 2015-08-25 Cardiome Pharma Corp. Synthetic process for aminocyclohexyl ether compounds
US8574259B2 (en) 2005-05-10 2013-11-05 Lifescreen Sciences Llc Intravascular filter with drug reservoir
US8344162B2 (en) 2005-06-15 2013-01-01 Cardiome Pharma Corp. Synthetic processes for the preparation of aminocyclohexyl ether compounds
US8618311B2 (en) 2005-06-15 2013-12-31 Cardiome Pharma Corp. Synthetic processes for the preparation of aminocyclohexyl ether compounds
US8080673B2 (en) 2005-06-15 2011-12-20 Cardiome Pharma Corp. Synthetic processes for the preparation of aminocyclohexyl ether compounds
US7754897B2 (en) 2005-06-15 2010-07-13 Cardiome Pharma Corp. Synthetic processes for the preparation of aminocyclohexyl ether compounds
WO2007015761A3 (en) * 2005-07-21 2009-04-16 Fmc Biopolymer As Medical devices coated with a fast dissolving biocompatible coating
US8257727B2 (en) 2005-07-21 2012-09-04 Fmc Biopolymer As Medical devices coated with a fast dissolving biocompatible coating
AU2006284657B2 (en) * 2005-08-31 2012-07-19 Abraxis Bioscience, Llc Compositions and methods for preparation of poorly water soluble drugs with increased stability
WO2007027941A3 (en) * 2005-08-31 2007-04-26 Abraxis Bioscience Inc Compositions and methods for preparation of poorly water soluble drugs with increased stability
TWI417114B (en) * 2005-08-31 2013-12-01 Abraxis Bioscience Llc Compositions and methods for preparation of poorly water soluble drugs with increased stability
US9308180B2 (en) 2005-08-31 2016-04-12 Abraxis Bioscience, Llc Compositions and methods for preparation of poorly water soluble drugs with increased stability
US7846445B2 (en) 2005-09-27 2010-12-07 Amunix Operating, Inc. Methods for production of unstructured recombinant polymers and uses thereof
US8492530B2 (en) 2005-09-27 2013-07-23 Amunix Operating Inc. Unstructured recombinant polymers and compositions comprising same
US9938331B2 (en) 2005-09-27 2018-04-10 Amunix Operating Inc. Biologically active proteins having increased in vivo and/or in vitro stability
US7855279B2 (en) 2005-09-27 2010-12-21 Amunix Operating, Inc. Unstructured recombinant polymers and uses thereof
US9855338B2 (en) 2005-12-05 2018-01-02 Nitto Denko Corporation Polyglutamate-amino acid conjugates and methods
US10507248B2 (en) 2005-12-19 2019-12-17 Pharmain Corporation Hydrophobic core carrier compositions for delivery of therapeutic agents, methods of making and using the same
US9737615B2 (en) 2005-12-19 2017-08-22 PharmalN Corporation Hydrophobic core carrier compositions for delivery of therapeutic agents, methods of making and using the same
WO2007124700A3 (en) * 2006-05-03 2007-12-21 I Q A A S Pharmaceutical composition containing taxane derivative destined for the preparation of an infusion solution, method of preparation thereof and use thereof
US8329853B2 (en) 2006-06-15 2012-12-11 Cell Therapeutics, Inc. Process for the preparation of poly-α-glutamic acid and derivatives thereof
EP2068956B1 (en) * 2006-06-26 2017-03-01 Boston Scientific Limited Medical devices for release of low solubility therapeutic agents
US10814013B2 (en) 2006-10-05 2020-10-27 The Board Of Regents Of The University Of Texas System Efficient synthesis of chelators for nuclear imaging and radiotherapy: compositions and applications
US10925977B2 (en) 2006-10-05 2021-02-23 Ceil>Point, LLC Efficient synthesis of chelators for nuclear imaging and radiotherapy: compositions and applications
WO2008062945A1 (en) * 2006-11-22 2008-05-29 Standard Sci-Tech Inc. Stent
CN101541268B (en) * 2006-11-22 2011-05-11 标准科学技术株式会社 Stent
US8937180B2 (en) 2006-11-30 2015-01-20 Nektar Therapeutics Method for preparing a polymer conjugate
US8354549B2 (en) 2006-11-30 2013-01-15 Nektar Therapeutics Method for preparing a polymer conjugate
US8541608B2 (en) 2006-11-30 2013-09-24 Nektar Therapeutics Method for preparing a polymer conjugate
US9610360B2 (en) 2007-01-24 2017-04-04 Ceruliean Pharma Inc. Polymer drug conjugates with tether groups for controlled drug delivery
RU2447443C2 (en) * 2007-03-06 2012-04-10 Селл Терапьютикс Инк. Method for determining amount of conjugated taxane in polyglutamic acid-taxane conjugates
US8318503B2 (en) 2007-03-06 2012-11-27 Cell Therapeutics, Inc. Method for determining the amount of conjugated taxane in polyglut acid-taxane conjugates
US9192697B2 (en) 2007-07-03 2015-11-24 Hemoteq Ag Balloon catheter for treating stenosis of body passages and for preventing threatening restenosis
US8518876B2 (en) 2007-08-03 2013-08-27 PharmalN Corporation Composition for long-acting peptide analogs
US9657078B2 (en) 2007-08-03 2017-05-23 Pharmain Corporation Composition for long-acting peptide analogs
US7960336B2 (en) 2007-08-03 2011-06-14 Pharmain Corporation Composition for long-acting peptide analogs
US9090664B2 (en) 2007-08-03 2015-07-28 Pharmain Corporation Composition for long-acting peptide analogs
US8933197B2 (en) 2007-08-15 2015-01-13 Amunix Operating Inc. Compositions comprising modified biologically active polypeptides
US8563527B2 (en) 2007-08-20 2013-10-22 Pharmain Corporation Oligonucleotide core carrier compositions for delivery of nucleic acid-containing therapeutic agents, methods of making and using the same
US8999930B2 (en) 2008-01-09 2015-04-07 Pharmain Corporation Soluble hydrophobic core carrier compositions for delivery of therapeutic agents, methods of making and using the same
US9562111B2 (en) 2008-01-09 2017-02-07 Pharmain Corporation Soluble hydrophobic core carrier compositions for delivery of therapeutic agents, methods of making and using the same
US8962566B2 (en) 2008-08-11 2015-02-24 Nektar Therapeutics Multi-arm polymeric alkanoate conjugates
US11672776B2 (en) 2008-08-11 2023-06-13 Nektar Therapeutics Multi-arm polymeric alkanoate conjugates
US9504755B2 (en) 2008-08-11 2016-11-29 Nektar Therapeutics Multi-arm polymeric alkanoate conjugates
US10039737B2 (en) 2008-08-11 2018-08-07 Nektar Therapeutics Multi-arm polymeric alkanoate conjugates
US9220790B2 (en) 2008-08-11 2015-12-29 Naktar Therapeutics Multi-arm polymeric alkanoate conjugates
US8637466B2 (en) 2008-08-11 2014-01-28 Nektar Therapeutics Multi-arm polymeric alkanoate conjugates
US9801873B2 (en) 2008-09-23 2017-10-31 Nektar Therapeutics Methods for treating humans having brain cancer
US10525051B2 (en) 2008-09-23 2020-01-07 Nektar Therapeutics Compositions and methods for achieving sustained therapeutic drug concentrations in a subject
US8906353B2 (en) 2008-09-23 2014-12-09 Nektar Therapeutics Compositions and methods for achieving sustained therapeutic drug concentrations in a subject
US9730820B2 (en) 2008-09-25 2017-08-15 Abbott Cardiovascular Systems Inc. Stent delivery system having a fibrous matrix covering with improved stent retention
US8491880B2 (en) 2008-12-10 2013-07-23 Mersana Therapeutics, Inc. Pharmaceutical formulations of biodegradable biocompatible camptothecin-polymer conjugates
US8673860B2 (en) 2009-02-03 2014-03-18 Amunix Operating Inc. Extended recombinant polypeptides and compositions comprising same
US10961287B2 (en) 2009-02-03 2021-03-30 Amunix Pharmaceuticals, Inc Extended recombinant polypeptides and compositions comprising same
US8680050B2 (en) 2009-02-03 2014-03-25 Amunix Operating Inc. Growth hormone polypeptides fused to extended recombinant polypeptides and methods of making and using same
US8957021B2 (en) 2009-02-03 2015-02-17 Amunix Operating Inc. Glucose-regulating polypeptides and methods of making and using same
US9168312B2 (en) 2009-02-03 2015-10-27 Amunix Operating Inc. Growth hormone polypeptides and methods of making and using same
US8716448B2 (en) 2009-02-03 2014-05-06 Amunix Operating Inc. Coagulation factor VII compositions and methods of making and using same
US8703717B2 (en) 2009-02-03 2014-04-22 Amunix Operating Inc. Growth hormone polypeptides and methods of making and using same
US9371369B2 (en) 2009-02-03 2016-06-21 Amunix Operating Inc. Extended recombinant polypeptides and compositions comprising same
US9926351B2 (en) 2009-02-03 2018-03-27 Amunix Operating Inc. Extended recombinant polypeptides and compositions comprising same
US10000543B2 (en) 2009-06-08 2018-06-19 Amunix Operating Inc. Glucose-regulating polypeptides and methods of making and using same
US9540430B2 (en) 2009-06-08 2017-01-10 Amunix Operating Inc. Glucose-regulating polypeptides and methods of making and using same
US9849188B2 (en) 2009-06-08 2017-12-26 Amunix Operating Inc. Growth hormone polypeptides and methods of making and using same
US10369256B2 (en) 2009-07-10 2019-08-06 Boston Scientific Scimed, Inc. Use of nanocrystals for drug delivery from a balloon
US11278648B2 (en) 2009-07-10 2022-03-22 Boston Scientific Scimed, Inc. Use of nanocrystals for drug delivery from a balloon
US10080821B2 (en) 2009-07-17 2018-09-25 Boston Scientific Scimed, Inc. Nucleation of drug delivery balloons to provide improved crystal size and density
US9758776B2 (en) 2009-08-24 2017-09-12 Amunix Operating Inc. Coagulation factor IX compositions and methods of making and using same
US9062299B2 (en) 2009-08-24 2015-06-23 Amunix Operating Inc. Coagulation factor IX compositions and methods of making and using same
US9376672B2 (en) 2009-08-24 2016-06-28 Amunix Operating Inc. Coagulation factor IX compositions and methods of making and using same
EP3372617A2 (en) 2010-04-02 2018-09-12 Amunix Operating Inc. Binding fusion proteins, binding fusion protein-drug conjugates, xten-drug conjugates and methods of making and using same
US10870874B2 (en) 2010-04-02 2020-12-22 Amunix Pharmaceuticals, Inc. Binding fusion proteins, binding fusion protein-drug conjugates, XTEN-drug conjugates and methods of making and using same
US9976166B2 (en) 2010-04-02 2018-05-22 Amunix Operating Inc. Binding fusion proteins, binding fusion protein-drug conjugates, XTEN-drug conjugates and methods of making and using same
US9249211B2 (en) 2010-04-02 2016-02-02 Amunix Operating Inc. Binding fusion proteins, binding fusion protein-drug conjugates, XTEN-drug conjugates and methods of making and using same
US8557961B2 (en) 2010-04-02 2013-10-15 Amunix Operating Inc. Alpha 1-antitrypsin compositions and methods of making and using same
US8889211B2 (en) 2010-09-02 2014-11-18 Boston Scientific Scimed, Inc. Coating process for drug delivery balloons using heat-induced rewrap memory
US10894087B2 (en) 2010-12-22 2021-01-19 Nektar Therapeutics Multi-arm polymeric prodrug conjugates of cabazitaxel-based compounds
US11813241B2 (en) 2010-12-22 2023-11-14 Nektar Therapeutics Multi-arm polymeric prodrug conjugates of taxane-based compounds
US10098865B2 (en) 2010-12-22 2018-10-16 Nektar Therapeutics Multi-arm polymeric prodrug conjugates of taxane-based compounds
US9056152B2 (en) 2011-08-25 2015-06-16 Boston Scientific Scimed, Inc. Medical device with crystalline drug coating
US10421798B2 (en) 2012-02-15 2019-09-24 Bioverativ Therapeutics Inc. Factor VIII compositions and methods of making and using same
US10370430B2 (en) 2012-02-15 2019-08-06 Bioverativ Therapeutics Inc. Recombinant factor VIII proteins
US11685771B2 (en) 2012-02-15 2023-06-27 Bioverativ Therapeutics Inc. Recombinant factor VIII proteins
US10953073B2 (en) 2012-02-27 2021-03-23 Amunix Pharmaceuticals, Inc. XTEN conjugate compositions and methods of making same
US10172953B2 (en) 2012-02-27 2019-01-08 Amunix Operating Inc. XTEN conjugate compositions and methods of making same
US11464871B2 (en) 2012-10-02 2022-10-11 Novartis Ag Methods and systems for polymer precipitation and generation of particles
US9993427B2 (en) 2013-03-14 2018-06-12 Biorest Ltd. Liposome formulation and manufacture
US11633357B2 (en) 2013-03-14 2023-04-25 Zuli Holdings, Ltd. Liposome formulation and manufacture
US10265269B2 (en) 2013-03-14 2019-04-23 Biorest Ltd. Liposome formulation and manufacture
US9901663B2 (en) 2013-05-06 2018-02-27 Abbott Cardiovascular Systems Inc. Hollow stent filled with a therapeutic agent formulation
CN103263675A (en) * 2013-05-16 2013-08-28 湘潭大学 Poly (epsilon-caprolactone) supported anti-tumor prodrug and preparation method thereof
US10548953B2 (en) 2013-08-14 2020-02-04 Bioverativ Therapeutics Inc. Factor VIII-XTEN fusions and uses thereof
US10745680B2 (en) 2015-08-03 2020-08-18 Bioverativ Therapeutics Inc. Factor IX fusion proteins and methods of making and using same
US11713358B2 (en) 2015-08-28 2023-08-01 Amunix Pharmaceuticals, Inc. Chimeric polypeptide assembly and methods of making and using the same
EP3917397A4 (en) * 2019-01-28 2022-10-26 Board of Regents, The University of Texas System Metal chelator combination therapy for the treatment of cancer

Also Published As

Publication number Publication date
IL126179A (en) 2003-04-10
AU735900B2 (en) 2001-07-19
HU226646B1 (en) 2009-05-28
EP1683520A2 (en) 2006-07-26
CA2250295C (en) 2008-12-30
ES2448467T3 (en) 2014-03-14
NZ332234A (en) 2000-06-23
EA199800817A1 (en) 1999-04-29
CN1217662A (en) 1999-05-26
ATE314843T1 (en) 2006-02-15
PT932399E (en) 2006-05-31
DE69735057T2 (en) 2006-08-31
EP0932399A4 (en) 2002-10-24
KR100561788B1 (en) 2006-09-20
EP1683520B1 (en) 2013-11-20
SI0932399T1 (en) 2006-10-31
PL328807A1 (en) 1999-02-15
JP3737518B2 (en) 2006-01-18
EA002400B1 (en) 2002-04-25
KR20000067033A (en) 2000-11-15
US6262107B1 (en) 2001-07-17
NO324461B1 (en) 2007-10-22
CN101028259A (en) 2007-09-05
DE69735057D1 (en) 2006-03-30
EP0932399A1 (en) 1999-08-04
IL126179A0 (en) 1999-05-09
NO984210D0 (en) 1998-09-11
UA68330C2 (en) 2004-08-16
DK0932399T3 (en) 2006-05-15
CA2250295A1 (en) 1997-09-18
PL189698B1 (en) 2005-09-30
ES2258790T3 (en) 2006-09-01
NO984210L (en) 1998-11-11
EP0932399B1 (en) 2006-01-04
HUP9903952A3 (en) 2001-06-28
CZ290898A3 (en) 1999-07-14
CZ297979B6 (en) 2007-05-16
US5977163A (en) 1999-11-02
JP2003063960A (en) 2003-03-05
NO332539B1 (en) 2012-10-15
NO20072562L (en) 1998-11-11
CN1304058C (en) 2007-03-14
BR9710646A (en) 2000-01-11
EP1683520A3 (en) 2009-11-18
AU2580697A (en) 1997-10-01
JP2000507930A (en) 2000-06-27
HUP9903952A2 (en) 2001-05-28

Similar Documents

Publication Publication Date Title
US5977163A (en) Water soluble paclitaxel prodrugs
US6884817B2 (en) Water soluble paclitaxel derivatives
KR100547931B1 (en) A composition comprising water soluble parclitaxel prodrugs and an implantable medical device comprising the same
Copie et al. Water soluble paclitaxel prodrugs
MXPA98007442A (en) Soluble paclitaxel profarmacos in a

Legal Events

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

Ref document number: 97194360.5

Country of ref document: CN

AK Designated states

Kind code of ref document: A1

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

AL Designated countries for regional patents

Kind code of ref document: A1

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

121 Ep: the epo has been informed by wipo that ep was designated in this application
ENP Entry into the national phase

Ref document number: 2250295

Country of ref document: CA

Ref document number: 2250295

Country of ref document: CA

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: PA/a/1998/007442

Country of ref document: MX

Ref document number: PV1998-2908

Country of ref document: CZ

WWE Wipo information: entry into national phase

Ref document number: 1019980711024

Country of ref document: KR

WWE Wipo information: entry into national phase

Ref document number: 332234

Country of ref document: NZ

WWE Wipo information: entry into national phase

Ref document number: 1997917512

Country of ref document: EP

Ref document number: 199800817

Country of ref document: EA

DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

WWP Wipo information: published in national office

Ref document number: PV1998-2908

Country of ref document: CZ

WWP Wipo information: published in national office

Ref document number: 1997917512

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 1019980711024

Country of ref document: KR

WWR Wipo information: refused in national office

Ref document number: 1019980711024

Country of ref document: KR

WWG Wipo information: grant in national office

Ref document number: 1997917512

Country of ref document: EP

WWG Wipo information: grant in national office

Ref document number: PV1998-2908

Country of ref document: CZ