US20010034363A1 - Water soluble paclitaxel derivatives - Google Patents

Water soluble paclitaxel derivatives Download PDF

Info

Publication number
US20010034363A1
US20010034363A1 US09/050,662 US5066298A US2001034363A1 US 20010034363 A1 US20010034363 A1 US 20010034363A1 US 5066298 A US5066298 A US 5066298A US 2001034363 A1 US2001034363 A1 US 2001034363A1
Authority
US
United States
Prior art keywords
paclitaxel
poly
composition
cancer
txl
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
US09/050,662
Other versions
US6441025B2 (en
Inventor
Chun Li
Sidney Wallace
Dong-Fang Yu
David J. Yang
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
PG TXL Co LP
Original Assignee
Individual
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from US08/815,104 external-priority patent/US5977163A/en
Application filed by Individual filed Critical Individual
Priority to US09/050,662 priority Critical patent/US6441025B2/en
Assigned to PG-TXL COMPANY, L.P. reassignment PG-TXL COMPANY, L.P. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: LI, CHUN, WALLACE, SIDNEY, YANG, DAVID J., YU, DONG-FANG
Priority to EP99916188A priority patent/EP1028756B1/en
Priority to CA2303338A priority patent/CA2303338C/en
Priority to AU34556/99A priority patent/AU3455699A/en
Priority to TW088104989A priority patent/TWI246922B/en
Priority to AT99916188T priority patent/ATE304867T1/en
Priority to MYPI99001207A priority patent/MY121807A/en
Priority to DK99916188T priority patent/DK1028756T3/en
Priority to EP05007529A priority patent/EP1598081A3/en
Priority to ES99916188T priority patent/ES2249888T3/en
Priority to TW093136237A priority patent/TWI255183B/en
Priority to PCT/US1999/006870 priority patent/WO1999049901A1/en
Priority to DE69927350T priority patent/DE69927350T2/en
Publication of US20010034363A1 publication Critical patent/US20010034363A1/en
Priority to US10/146,809 priority patent/US6884817B2/en
Priority to US10/153,818 priority patent/US6515017B1/en
Publication of US6441025B2 publication Critical patent/US6441025B2/en
Application granted granted Critical
Priority to US10/243,079 priority patent/US20030114397A1/en
Priority to US10/243,045 priority patent/US20030114518A1/en
Priority to US10/243,080 priority patent/US20030114363A1/en
Priority to US10/243,046 priority patent/US20030113335A1/en
Priority to US10/282,490 priority patent/US7135496B2/en
Priority to US10/282,570 priority patent/US20030134793A1/en
Priority to US10/298,349 priority patent/US20030130170A1/en
Priority to US10/298,327 priority patent/US20030130178A1/en
Priority to US10/298,375 priority patent/US20030130341A1/en
Priority to US10/300,031 priority patent/US6730699B2/en
Priority to US10/310,331 priority patent/US20030147807A1/en
Priority to US10/310,511 priority patent/US20030124055A1/en
Priority to US10/354,431 priority patent/US7060724B2/en
Priority to US10/620,238 priority patent/US20040018960A1/en
Priority to US10/826,302 priority patent/US20040198638A1/en
Priority to US11/253,810 priority patent/US20060111273A1/en
Priority to US11/285,392 priority patent/US20060135404A1/en
Priority to US11/513,390 priority patent/US7384977B2/en
Priority to US11/927,392 priority patent/US20080153865A1/en
Anticipated expiration legal-status Critical
Expired - Lifetime legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K41/00Medicinal preparations obtained by treating materials with wave energy or particle radiation ; Therapies using these preparations
    • A61K41/0038Radiosensitizing, i.e. administration of pharmaceutical agents that enhance the effect of radiotherapy
    • 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
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/4353Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/4375Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems the heterocyclic ring system containing a six-membered ring having nitrogen as a ring heteroatom, e.g. quinolizines, naphthyridines, berberine, vincamine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/04Peptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
    • A61K38/12Cyclic peptides, e.g. bacitracins; Polymyxins; Gramicidins S, C; Tyrocidins A, B or C
    • A61K38/13Cyclosporins
    • 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
    • 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
    • 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
    • A61K51/06Macromolecular compounds, carriers being organic macromolecular compounds, i.e. organic oligomeric, polymeric, dendrimeric molecules
    • A61K51/065Macromolecular compounds, carriers being organic macromolecular compounds, i.e. organic oligomeric, polymeric, dendrimeric molecules conjugates with carriers being macromolecules
    • 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
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/04Antineoplastic agents specific for metastasis
    • CCHEMISTRY; METALLURGY
    • C08ORGANIC MACROMOLECULAR COMPOUNDS; THEIR PREPARATION OR CHEMICAL WORKING-UP; COMPOSITIONS BASED THEREON
    • C08GMACROMOLECULAR COMPOUNDS OBTAINED OTHERWISE THAN BY REACTIONS ONLY INVOLVING UNSATURATED CARBON-TO-CARBON BONDS
    • C08G73/00Macromolecular compounds obtained by reactions forming a linkage containing nitrogen with or without oxygen or carbon in the main chain of the macromolecule, not provided for in groups C08G12/00 - C08G71/00
    • C08G73/06Polycondensates having nitrogen-containing heterocyclic rings in the main chain of the macromolecule
    • C08G73/10Polyimides; Polyester-imides; Polyamide-imides; Polyamide acids or similar polyimide precursors
    • C08G73/1092Polysuccinimides
    • 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 (TaxolTM) and docetaxel (Taxotere), in particular making paclitaxel water soluble by conjugating the drug to water soluble moieties.
  • anticancer agents such as paclitaxel (TaxolTM) and docetaxel (Taxotere)
  • Paclitaxel an anti-microtubule agent extracted from the needles and bark of the Pacific yew tree, Taxus brevifolia
  • 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.
  • 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
  • Dehydrated alcohol 50% v/v
  • Paclitaxel (TaxolTM) has shown significant activity in human cancers, including breast, ovarian, non-small cell lung, and head and neck cancers (Rowinsky and Donehower, 1995). It has also shown significant activity in patients with advanced breast cancer who had been treated with multiple chemotherapeutic agents (Foa et al., 1994).
  • paclitaxel As with most chemotherapeutic agents, however, the maximum tolerated dose of paclitaxel is limited by toxicity. In humans, paclitaxel's major toxic effect at doses of 100-250 mg/m 2 is granulocytopenia (Holmes et al., 1995); symptomatic peripheral neuropathy is its principal nonhematologic toxicity (Rowinsky et al., 1993).
  • 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 Several toxic effects have been attributed to Cremophor, including vasodilatation, dyspnea, and hypotension. This vehicle has also been shown to cause serious hypersensitivity in laboratory animals and humans (Weiss et al., 1990).
  • 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 particulate 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.
  • protaxols novel type of prodrug termed “protaxols”. These compounds possess greater aqueous solubility and are converted to paclitaxel as the active drug through an intramolecular hydrolysis mechanism. However, no in vivo data on the antitumor activity of protaxols are yet available. Greenwald et al. reported the synthesis of highly water-soluble 2′ and 7-polyethylene glycol esters of paclitaxel (Greenwald et al., 1994).
  • PEG polyethylene glycol
  • 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 angioplasty and stenting.
  • compositions comprising a chemotherapeutic and/or antiangiogenic drug, such as paclitaxel, docetaxel, or other taxoid conjugated to a water soluble polymer such as a water soluble polyamino acid, or to a water soluble metal chelator. It is a further embodiment of the present invention that a composition comprising a conjugate of paclitaxel and poly-glutamic acid has surprising antitumor activity in animal models, and further that this composition is demonstrated herein to be a new species of taxane that has pharmaceutical properties different from that of paclitaxel.
  • a chemotherapeutic and/or antiangiogenic drug such as paclitaxel, docetaxel, or other taxoid conjugated to a water soluble polymer such as a water soluble polyamino acid, or to a water soluble metal chelator.
  • 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, docetaxel, or other taxoid 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 improved efficacy and controlled release so that tumors are shown herein to be eradicated in animal models after a single intravenous administration, as well as providing a novel taxane.
  • Poly-(l-glutamic acid) conjugated paclitaxel is shown in the examples hereinbelow to have a novel drug activity, in addition to having improved the delivery to the tumor and providing a 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 paclitaxel, tamoxifen, Taxotere, etopside, teniposide, fludarabine, doxorubicin, daunomycin, emodin, 5-fluorouracil, FUDR, estradiol, camptothecin, retinoids, verapamil, epothilones cyclosporin, and other taxoids.
  • those agents with a free hydroxyl group would be conjugated to the polymers by similar chemical reactions as described herein for paclitaxel.
  • conjugated to a water soluble polymer means the covalent bonding of the drug to the polymer or chelator.
  • the water soluble conjugates of the present invention may be administered in conjunction with other drugs, including other anti-tumor or anti-cancer drugs.
  • other drugs including other anti-tumor or anti-cancer drugs.
  • the water soluble paclitaxel, docetaxel, or other taxoid, or in preferred embodiments the poly-(l-glutamic) acid conjugated paclitaxel (PG-TXL), of the present invention may, in certain types of treatment, be combined with a platinum drug, an antitumor agent such as doxorubicin or daunorubicin, for example, or other drugs that are used in combination with TaxolTM or combined with external or internal irradiation.
  • an antitumor agent such as doxorubicin or daunorubicin
  • 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.
  • the water soluble polymers such as, in preferred embodiments PG-TXL, may have pharmacological properties different from non-conjugated drugs (i.e. paclitaxel). Additionally, polymer-drug conjugates may act as drug depots for sustained release, producing prolonged drug exposure to tumor cells.
  • water soluble polymers e.g., water soluble polyamino acids
  • 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).
  • SMANCS Session Management System
  • HPMA-Dox in the United Kingdom
  • 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 a preferred taxane is a chemical of the general chemical formula, C 47 H 5 , NO 14 , including [2aR-[2a ⁇ ,4 ⁇ ,4 ⁇ ,6 ⁇ ,9 ⁇ ( ⁇ R*, ⁇ S*), 11 ⁇ , 12 ⁇ , 12a ⁇ , 12 ⁇ ,]]- ⁇ -(Benzoylamino)- ⁇ -hydroxybenzene propanoic 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-7,11
  • 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 or taxane 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 metals.
  • Preferred radionuclides include, but are not limited to 67 Ga, 68 Ga, 111 In, 99m Tc, 90 Y, 114m Sn and 193m Pt.
  • Preferred water soluble chelators to be used in the practice of the present invention include, but are not limited to, diethylenetriaminepentaacetic acid (DTPA), ethylenediaminetetraacetic acid (EDTA), 1,4,7,1 0-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 1-(p-aminobenzyl)-DTPA, 1,6-diamino hexane-N,N,N′,N′-tetraacetic acid, DPDP, and ethylenebis (oxyethylenenitrilo)-tetraacetic acid, with DTPA being
  • the paclitaxel, docetaxel, or other taxoid 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, docetaxel, or other taxoid.
  • Poly-glutamic acid (PG) is one polymer that offers several advantages in the present invention. First, it contains a large number of side chain carboxyl functional groups for drug attachment. Second, PG can be readily degraded by lysosomal enzymes to its nontoxic basic component, 1-glutamic acid, d-glutamic acid and di-glutamic acid.
  • Preferred polymers include, but are not limited to poly(l-glutamic acid), poly(d-glutamic acid), poly(dl-glutamic acid), poly(l-aspartic acid), poly(d-aspartic acid), poly(dl-aspartic acid), poly(l-lysine), poly(d-lysine), poly(dl-lysine), copolymers of the above listed polyamino acids with polyethylene glycol, polycaprolactone, polyglycolic acid and polylactic acid, as well as poly(2-hydroxyethyl 1-glutamine), chitosan, carboxymethyl dextran, hyaluronic acid, human serum albumin and alginic acid, with poly-glutamic acids being particularly preferred.
  • the polymers of the present invention preferably have a molecular weight of about 1,000, about 2,000, about 3,000, about 4,000, about 5,000, about 6,000, about 7,000, about 8,000, about 9,000, about 10,000, about 11,000, about 12,000, about 13,000, about 14,000, about 15,000, about 16,000, about 17,000, about 18,000, about 19,000, about 20,000, about 21,000, about 22,000, about 23,000, about 24,000, about 25,000, about 26,000, about 27,000, about 28,000, about 29,000, about 30,000, about 31,000, about 32,000, about 33,000, about 34,000, about 35,000, about 36,000, about 37,000, about 38,000, about 39,000, about 40,000, about 41,000, about 42,000, about 43,000, about 44,000, about 45,000, about 46,000, about 47,000, about 48,000, about 49,000, to about 50,000 kd.
  • the polymers of the present invention preferably have a molecular weight of about 51,000, about 52,000, about 53,000, about 54,000, about 55,000, about 56,000, about 57,000, about 58,000, about 59,000, about 60,000, about 61,000, about 62,000, about 63,000, about 64,000, about 65,000, about 66,000, about 67,000, about 68,000, about 69,000, about 70,000, about 71,000, about 72,000, about 73,000, about 74,000, about 75,000, about 76,000, about 77,000, about 78,000, about 79,000, about 80,000, about 81,000, about 82,000, about 83,000, about 84,000, about 85,000, about 86,000, about 87,000, about 88,000, about 89,000, about 90,000, about 91,000, about 92,000, about 93,000, about 94,000, about 95,000, about 96,000, about 97,000, about 98,000, about 99,000, to about 100,000 kd.
  • the ranges of molecular weights for the polymers are preferably of about 91,000, about 92,000, about 93,000, about 9
  • a composition of the invention such as PG-TXL may also be conjugated to a second lipophilic or poorly soluble antitumor agent such as camptothecin, epothilone, cisplatin, melphalan, Taxotere, etoposide, teniposide, fludarabine, verapamil, or cyclosporin, for example, or even to water soluble agents such as 5 fluorouracil (5 FU) or fluorodeoxyuridine (FUDR), doxorubicin or daunomycin.
  • a second lipophilic or poorly soluble antitumor agent such as camptothecin, epothilone, cisplatin, melphalan, Taxotere, etoposide, teniposide, fludarabine, verapamil, or cyclosporin, for example, or even to water soluble agents such as 5 fluorouracil (5 FU) or fluorodeoxyuridine (FUDR), dox
  • compositions of the present invention may be dispersed in a pharmaceutically acceptable carrier solution as described below.
  • a pharmaceutically acceptable carrier 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, docetaxel, or other taxoid conjugated to a high molecular weight water soluble polymer or to a chelator.
  • a chemotherapeutic or anti-cancer drug such as paclitaxel, docetaxel, or other taxoid conjugated to a high molecular weight water soluble polymer or to a chelator.
  • the pharmaceutical composition may include polyethylene glycol, poly-glutamic acids, poly-aspartic acids, poly-lysine, 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.
  • the present invention may be described as a method of determining the uptake of a chemotherapeutic drug such as paclitaxel, docetaxel, or other taxoid 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, docetaxel, or other taxoid 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.
  • a chemotherapeutic drug such as paclitaxel, docetaxel, or other taxoid conjugated to a water soluble polymer or chelator
  • compositions comprise paclitaxel, docetaxel, or other taxoid conjugated to a water soluble polyamino acids, including but not limited to poly (l-aspartic acid), poly (d-aspartic acid), or poly (dl-aspartic acid), poly (l-lysine acid), poly (d-lysine acid), or poly (dl-lysine acid), and more preferably to poly (l-glutamic acid), poly (d-glutamic acid), or poly (dl-glutamic acid).
  • a water soluble polyamino acids including but not limited to poly (l-aspartic acid), poly (d-aspartic acid), or poly (dl-aspartic acid), poly (l-lysine acid), poly (d-lysine acid), or poly (dl-lysine acid), and more preferably to poly (l-glutamic acid), poly (d-glutamic acid), or poly (dl-glutamic 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, head and neck cancer.
  • the compositions of the invention may also be used against gastric cancer, prostate cancer, colon cancer, leukemia, or Kaposi's Sarcoma.
  • the term “treating” cancer is understood as meaning any medical management of a subject having a tumor. The term would encompass any inhibition of tumor growth or metastasis, or any attempt to inhibit, slow or abrogate tumor growth or metastasis.
  • the method includes killing a cancer cell by non-apoptotic as well as apoptotic mechanisms of cell death.
  • 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, by methods that include but are not limited to bolus injection or infusion, as well as intraarterial, intravenous, intraperitoneal, or intratumoral administration of the drug.
  • 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 polymer, with polyamino acids being preferred and poly-glutamic acid being more preferred.
  • a composition comprising paclitaxel or docetaxel conjugated to polymer, with polyamino acids being preferred and poly-glutamic acid being more preferred.
  • Cremophor formulation U.S. Pat. No. 5,583,153, incorporated herein by reference.
  • compositions of the present invention are expected to be as effective as paclitaxel against rheumatoid arthritis.
  • Paclitaxel is an antiangiogenic agent.
  • Rheumatoid arthritis creates a collection of newly formed vessels which erode the adjacent joints.
  • the taxoid or taxane 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 other anti-cancer drugs, such as methotrexate.
  • AGM-1470 an angiogenesis inhibitor
  • anti-cancer drugs such as methotrexate.
  • water soluble paclitaxels and docetaxels of the present invention will find a variety of applications beyond direct parenteral administration (WO 9625176, incorporated herein by reference).
  • 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 stent 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-glutamic acid or other water soluble poly-amino acids.
  • the subject may be a coronary bypass, vascular surgery, organ transplant or coronary or any other arterial angioplasty patient, for example, and the composition may be administered directly, intravenously, or even coated on a stent to be 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 poly-glutamic acids or water soluble polyamino 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 during or after balloon angioplasty and the coating is effective to inhibit restenosis.
  • the invention may be described as a composition comprising poly-glutamic acids conjugated to the 2′ or 7 hydroxyl or both of paclitaxel, docetaxel, or other taxoids, or even a composition comprising water soluble polyamino acids conjugated to the 2′ or 7 hydroxyl or both of paclitaxel, docetaxel, or other taxoids.
  • a poly-glutamic acid or “poly-glutamic acids” include poly (l-glutamic acid), poly (d-glutamic acid) and poly (dl-glutamic acid), the terms “a poly-aspartic acid” or “poly-aspartic acids” include poly (l-aspartic acid), poly (d-aspartic acid), poly (dl-aspartic acid), the terms “a poly-lysine” or “poly-lysine” include poly (l-lysine), poly (d-lysine), poly (dl-lysine), and the terms “a water soluble polyamino acid”, “water soluble polyamino acids”, or “water soluble polymer of amino acids” include, but are not limited to, poly-glutamic acid, poly-aspartic acid, poly-lysine, and amino acid chains comprising mixtures of glutamic acid, aspartic acid, and/or lysine.
  • a water soluble polyamino acid examples include amino acid chains comprising combinations of glutamic acid and/or aspartic acid and/or lysine, of either d and/or l isomer conformation.
  • such a “water soluble polyamino acid” contains one or more glutamic acid, aspartic acid, and/or lysine residues.
  • Such “water soluble polyamino acids” may also comprise any natural, modified, or unusual amino acid described herein, as long as the majority of residues, i.e. greater than 50%, comprise glutamic acid and/or aspartic acid and/or lysine.
  • a water soluble polymer of amino acids that contains more than one different type of amino acid residue is sometimes referred to herein as a “co-polymer”.
  • various substitutions of naturally occurring, unussual, or chemically modified amino acids may be made in the amino acid composition of the “water soluble polyamino acids”, and particularly in “poly-glutamic acids”, to produce a taxoid-polyamino acid conjugate of the present invention and still obtain molecules having like or otherwise desirable characteristics of solubility and/or therapeutic efficacy.
  • a polyamino acid such as poly-glutamic acid, poly-aspartic acid, poly-lysine, or water soluble amino acids chain or polymer comprising a mixture of glutamic acid, aspartic acid, and/or lysine, may, at the lower end of the amino acid substitution range, have about 1, about 2, about 3, about 4, about 5, about 6, about 7, about 8, about 9, about 10, about 12, about 13, about 14, about 15, about 16, about 17, about 18, about 19, about 20, about 21, about 22, about 23, about 24, or about 25 or more glutamic acid, aspartic acid, or lysine, residues, respectively, substituted by any of the naturally occurring, modified, or unusual amino acids described herein.
  • a polyamino acid such as poly-glutamic acid, poly-aspartic acid, poly-lysine, or a poly-amino acid chain comprising a mixture of some or all of these three amino acids may, at the lower end, have about 1%, about 2%, about 3%, about 4%, about 5%, about 6%, about 7%, about 8%, about 9%, about 10%, about 11%, about 12%, about 13%, about 14%, about 15%, about 16%, about 17%, about 18%, about 19%, about 20%, about 21%, about 22%, about 23%, about 24%, to about 25% or more glutamic acid, aspartic acid, or lysine residues, respectively, substituted by any of the naturally occurring, modified, or unusual amino acids described herein.
  • a polyamino acid such as poly-glutamic acid, poly-aspartic acid, or poly-lysine may, at the high end of the amino acid substitution range, have about 25%, about 26%, about 27%, about 28%, about 29%, about 30%, about 31%, about 32%, about 33%, about 34%, about 35%, about 36%, about 37%, about 38%, about 39%, about 40%, about 41%, about 42%, about 43%, about 44%, about 45%, about 46%, about 47%, about 48%, about 49%, to about 50% or so of the glutamic acid, aspartic acid, or lysine residues, respectively, substituted by any of the naturally occurring, modified, or unusual amino acids described herein, as long as the majority of residues comprise glutamic acid and/or aspartic acid and/or lysine.
  • amino acid substitution of the various water soluble amino acid polymers residues with a hydrophilicity index of +1 or
  • the amount of anti-tumor drug conjugated per water soluble polymer can vary.
  • such a composition may comprise from about 1%, about 2%, about 3%, about 4%, about 5%, about 6%, about 7%, about 8%, about 9%, or about 10%, about 11%, about 12%, about 13%, about 14%, about 15%, about 16%, about 17%, about 18%, about 19%, about 20%, about 21% about 22%, about 23%, about 24%, to about 25% (w/w) antitumor drug relative to the mass of the conjugate.
  • such a composition may comprise from about 26%, about 27%, about 28%, about 29%, about 30%, about 31% about 32%, about 33%, about 34%, about 35%, about 36%, about 37%, about 38%, about 39%, to about 40% or more (w/w) antitumor drug relative to the mass of the conjugate.
  • Preferred anti-tumor drugs include paclitaxel, docetaxel, or other taxoids
  • preferred water soluble polymers include water soluble amino acid polymers.
  • the number of molecules of anti-tumor drug conjugated per molecule of water soluble polymer can vary. At the lower end, such a composition may comprise from about 1, about 2, about 3, about 4, about 5, about 6, about 7, about 8, about 9, about 10, about 11, about 12, about 13, about 14, about 15, about 16, about 17, about 18, about 19, to about 20 or more molecules of antitumor drug per molecule of water soluble polymer.
  • such a composition may comprise from about 21, about 22, about 23, about 24, about 25, about 26, about 27, about 28, about 29, about 30, about 31, about 32, about 33, about 34, about 35, about 36, about 37, about 38, about 39, about 40, about 41, about 42, about 43, about 44, about 45, about 46, about 47, about 48, about 49, about 50, about 51, about 52, about 53, about 54, about 55, about 56, about 57, about 58, about 59, about 60 about 61, about 62, about 63, about 64, about 65, about 66, about 67, about 68, about 69, about 70, about 71, about 72, about 73, about 74, to about 75 or more molecules or more of antitumor drug per molecule of water soluble polymer.
  • Preferred anti-tumor drugs include paclitaxel, docetaxel, or other taxoids
  • preferred water soluble polymers include water soluble amino acid polymers.
  • the preferred number of anti-tumor drug molecules conjugated per molecule of water soluble polymer is about 7 molecules of antitumor drug per molecule of water soluble polymer.
  • Biological functional equivalents Water soluble amino acid polymers with various substitutions of residues conjugated to paclitaxel, docetaxel, or other taxoids are referred to as “biological functional equivalents”. These “biologically functional equivalents” are part of the definition of “water soluble polyamino acids” that are conjugated to taxoids, and may be identified by the assays described herein as well as any applicable assay that is known to those of skill in the art to measure improved aqueous solubility relative to the unconjugated taxoid or taxoids used to produce the particular water soluble amino acid polymer-taxoid composition.
  • biological functional equivalents of water soluble amino acid-taxoid polymers may be further identified by improved anti-tumor cell activity, relative to the anti-tumor cell activity of the unconjugated water soluble amino acid polymer used to produce the particular water soluble amino acid polymer-taxoid composition by the assays described herein as well as any applicable assay that is known to those of skill in the art.
  • biologically functional equivalents as used herein to describe this aspect of the invention is further described in the detailed description of the invention.
  • FIG. 1A Chemical structure of paclitaxel, PEG-paclitaxel and DTPA-paclitaxel.
  • FIG. 1B Chemical structure and reaction scheme for production of PG-TXL.
  • 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 111 In-DTPA-paclitaxel and 111 In-DTPA. Arrow indicates the tumor.
  • FIG. 6 Hydrolytic degradation of PG-TXL as determined in PBS as a function of time at different pH levels. - ⁇ - represents percent paclitaxel released, -O- represents metabolite-1 produced.
  • FIG. 7A Anti-tumor effect of PG-TXL against syngeneic OCA-I ovarian carcinoma tumor in female C3Hf/Kam mice. Drugs were injected intraveneously in a single dose. Data are presented as mean ⁇ standard deviation of tumor volumes.
  • FIG. 7B Anti-tumor effect of PG-TXL against 13762F tumor in female rats.
  • FIG. 7C The antitumor effect of PG-TXL on mice bearing MCa-4 mammary carcinoma tumors.
  • - ⁇ - represents the response to a single i.v. dose of saline
  • - ⁇ - represents the response to a single i.v. dose of PG (0.6 g/kg)
  • - ⁇ - represents response to PG-TXL (40 mg/kg)
  • - ⁇ - represents response to PG-TXL (60 mg equiv. paclitaxel/kg)
  • - - represents response to PG-TXL (120 mg/kg).
  • FIG. 7D The antitumor effect of PG-TXL against soft-tissue sarcoma tumor (FSa-II) in mice.
  • - ⁇ - 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)
  • - - represents response to paclitaxel (80 mg/kg)
  • - ⁇ - represents response to PG-TXL (160 mg equiv. paclitaxel/kg).
  • FIG. 7E The antitumor effect of PG-TXL against syngeneic hepatocarcinoma tumor (HCa-I) in mice.
  • - ⁇ - 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)
  • - - represents response to PG-TXL (80 mg/kg)
  • - ⁇ - represents response to PG-TXL (160 mg equiv. paclitaxel/kg).
  • FIG. 8 Release profile of paclitaxel from PEG-paclitaxel in phosphate buffer (pH 7.4). Release profiles of paclitaxel (- ⁇ -); from PEG-paclitaxel (- -) at pH 7.4 is shown.
  • FIG. 9 Antitumor effect of PEG-paclitaxel on MCa-4 mammary tumors.
  • - ⁇ - 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
  • - - represents a single dose of 40 mg/kg body weight of paclitaxel
  • - ⁇ - represents PEG-paclitaxel at 40 mg equiv. paclitaxel/kg body weight.
  • FIG. 10 Tubulin polymerization assays performed at 32° C. in the presence of 1.0 mM GTP and 1.0 mg/ml of tubulin.
  • - - represents paclitaxel (1.0 EM)
  • - ⁇ - represents PG-TXL (10 ⁇ M equivalent paclitaxel) incubated in PBS (pH 7.4) at 37° C. for 3 days
  • - - represents freshly dissolved PG-TXL.
  • FIG. 11 Plasma clearance of radioactivity following an i.v. injection of PG-[ 3 H]paclitaxel and [ 3 H]paclitaxel in C3Hf/Kam mice.
  • - - represents PG-TXL radioactivity after injection of 6 ⁇ Ci of radiolabeled PG-[ 3 H]paclitaxel (20 mg equivalent paclitaxel/kg)
  • - ⁇ - represents paclitaxel radioactivity after injection of 6 ⁇ Ci of radiolabeled [ 3 H]paclitaxel (20 mg equivalent paclitaxel/kg)
  • - - represents “Paclitaxel” radioactivity released from injected PG-[ 3 H]paclitaxel.
  • FIG. 12A Time-dependent OCA-1 tumor content of radioactivity following injection of either PG-[ 3 H]paclitaxel and [ 3 H]paclitaxel into mice.
  • Open bars represents PG-TXL radioactivity after injection of 6 ⁇ Ci of radiolabeled PG-[ 3 H]paclitaxel (20 mg equivalent paclitaxel/kg)
  • filled bars represents paclitaxel radioactivity after injection of 6 ⁇ Ci of radiolabeled [ 3 H]paclitaxel (20 mg equivalent paclitaxel/kg).
  • FIG. 12B Conversion of PG-[ 3 H]paclitaxel to [ 3 H]paclitaxel within OCA-1 tumor.
  • Total radioactivity measured after injection of 6 ⁇ Ci of radiolabeled PG-[ 3 H]paclitaxel is shown in open bars
  • “Paclitaxel” derived radioactivity released from injected PG-[ 3 H]paclitaxel is shown in solid bars.
  • FIG. 13 Kinetics of apoptosis in OCA-1 tumors after a single i.v. dose of 160 mg equiv. paclitaxel/kg of PG-TXL (MTD) and 80 mg/kg paclitaxel (MTD).
  • - ⁇ - represents the response to a single i.v. dose of PG-TXL (160 mg equiv. paclitaxel/kg MTD)
  • - ⁇ - represents response to paclitaxel (80 mg paclitaxel/kg MTD).
  • FIG. 14 Survival of nude mice with human ovarian cancer cells (SKOV3ip1) treated with PG-TXL.
  • Five days after tumor injection the mice were injected i.v. with the PG-paclitaxel (PG-TXL), or PG control. Injections of PG-TXL were administered every seven days ( ⁇ ) in the 120 mg/kg group, but not the 160 mg/kg group.
  • - ⁇ - represents untreated mice.
  • - ⁇ - represents the response to multiple i.v. doses of PG.
  • - ⁇ - represents the response to an i.v. dose of PG-TXL (120 mg equiv. paclitaxel/kg)
  • - ⁇ - represents the response to an i.v. dose of PG-TXL (160 mg equiv. paclitaxel/kg).
  • FIG. 15 Chemical structure and reaction scheme for production of glutamic acid containing polyamino acids.
  • 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 (l-glutamic acid) conjugated paclitaxel (PG-TXL) 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 maximum tolerated dose
  • PG-TXL is a novel species of taxane that is pharmacologically distinct from previous paclitaxel or TaxolTM preparations.
  • the distribution of PG-TXL within plasma is distinct from free paclitaxel. While paclitaxel remains in the plasma of mice for an extremely short time, PG-TXL appears to remain for a much longer period. This is contemplated to offer a distinct advantage in that prolonged exposure of tumors to the drug may result in an enhanced response.
  • the rate of conversion of PG-TXL to paclitaxel is slow, with less than 1% of the radioactivity from radiolabeled PG-TXL being recovered as radioactive paclitaxel within 48 h after injection of the paclitaxel-polymer complex.
  • the novel drug, PG-TXL may produce death within tumor cells in a manner which is not simply due to the gradual release of paclitaxel itself.
  • Another aspect of the present invention is the inclusion of molecules in the polymeric composition that are effective to target the therapeutic composition to a disease or tumor site or to a particular organ or tissue.
  • Many of such targeting molecules are known in the art and may be conjugated to the water soluble anti-tumor compositions of the present invention.
  • Examples of such molecules or agents would include, but not be limited to antibodies such as anti-tumor antibodies; anti-cell receptor antibodies; tissue specific antibodies; hormonal agents such as octreotide, estradiol and tamoxifen; growth factors; cell surface receptor ligands; enzymes; hypoxic agents such as misonidazole and erythronitroimidazole; and antiangiogenic agents.
  • composition of the present invention is DTPA-paclitaxel, 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.
  • 111 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 that also have different antitumor properties than unmodified paclitaxel.
  • 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 predict the efficacy of an anti-cancer treatment. This information may be helpful in guiding the practitioner in the selection of patients to undergo chelator-paclitaxel therapy.
  • SPECT single photon emission computer tomography
  • PET positron emission tomography
  • the paclitaxel may be rendered water-soluble in many ways: i.e. 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., 111 In, 90 Y, 166 Ho, 68 Ga, 99m Tc) for nuclear imaging and/or for radiotherapy studies.
  • radionuclides e.g., 111 In, 90 Y, 166 Ho, 68 Ga, 99m Tc
  • paclitaxel polyethylene glycol-paclitaxel
  • PEG-paclitaxel polyethylene glycol-paclitaxel
  • PG-TXL poly-glutamic acid-paclitaxel conjugate
  • DTPA-paclitaxel diethylenetriaminepentaacetic acid-paclitaxel
  • 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.
  • DTPA-paclitaxel or other paclitaxel-chelating agents will be useful as diagnostic agents which, when labeled with radionuclides such as 111 In or 99m Tc, may be used as radiotracers to detect certain tumors in combination with nuclear imaging techniques. It is understood that in addition to paclitaxel (TaxolTM) 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 present invention.
  • amino acids may be substituted for other amino acids in a polyamino acid structure, including water soluble amino acid polymers such as poly-glutamic acid, poly-aspartic acid, or poly-lysine, without appreciable loss of interactive binding capacity with structures such as, for example, a chemotherapeutic and/or antiangiogenic drug, such as paclitaxel or docetaxel, or such like.
  • a chemotherapeutic and/or antiangiogenic drug such as paclitaxel or docetaxel, or such like.
  • amino acid substitutions in a water soluble polyamino acid conjugated to a chemotherapeutic and/or antiangiogenic drug such as paclitaxel or docetaxel, or such like, as exemplified by but not limited to PG-TXL, may be made and still maintain part or all of the novel pharmacological properties disclosed herein. Since it is the interactive capacity and nature of a protein that defines that protein's biological functional activity, certain amino acid sequence substitutions can be made in a polyamino acid sequence and nevertheless obtain a polyamino acid with like (agonistic) properties. It is thus contemplated by the inventors that various changes may be made in the sequence of the water soluble polyamino acids of a drug conjugate, such as, but not limited to PG-TXL, without appreciable loss of their biological utility or activity.
  • Biologically functional equivalent of a water soluble polyamino acid is the concept that there is a limit to the number of changes that may be made within a portion of the molecule and still result in a molecule with an acceptable level of equivalent biological activity.
  • Biologically functional equivalent of a water soluble polyamino acids are thus defined herein as those water soluble polyamino acids in which certain, not most or all, of the amino acids may be substituted by non-water soluble amino acids, whether natural, unusual, or chemically modified.
  • Amino acid substitutions are generally based on the relative similarity of the amino acid side-chain substituents, for example, their hydrophobicity, hydrophilicity, charge, size, and the like.
  • An analysis of the size, shape and type of the amino acid side-chain substituents reveals that arginine, lysine and histidine are all positively charged residues; that alanine, glycine and serine are all a similar size; and that phenylalanine, tryptophan and tyrosine all have a generally similar shape.
  • arginine, lysine and histidine; alanine, glycine and serine; and phenylalanine, tryptophan and tyrosine; are defined herein as biologically functional equivalents.
  • hydropathic index of amino acids may be considered.
  • Each amino acid has been assigned a hydropathic index on the basis of their hydrophobicity and charge characteristics, these are: isoleucine (+4.5); valine (+4.2); leucine (+3.8); phenylalanine (+2.8); cysteine/cystine (+2.5); methionine (+1.9); alanine (+1.8); glycine ( ⁇ 0.4); threonine ( ⁇ 0.7); serine ( ⁇ 0.8); tryptophan ( ⁇ 0.9); tyrosine ( ⁇ 1.3); proline ( ⁇ 1.6); histidine ( ⁇ 3.2); glutamate ( ⁇ 3.5); glutamine ( ⁇ 3.5); aspartate ( ⁇ 3.5); asparagine ( ⁇ 3.5); lysine ( ⁇ 3.9); and arginine ( ⁇ 4.5).
  • hydrophilicity In making changes based upon similar hydrophilicity values, the substitution of amino acids whose hydrophilicity values are within ⁇ 2 is preferred, those which are within ⁇ 1 are particularly preferred, and those within ⁇ 0.5 are even more particularly preferred.
  • arginine, lysine, aspartic acid, and glutamic acid are defined herein as biologically functional equivalents, particularly in water soluble amino acid polymers.
  • water soluble polyamino acid-chemotherapeutic and/or antiangiogenic drug compounds described herein such as paclitaxel or docetaxel conjugated to a water soluble amino acid, or such like, as exemplified by, but not limited to PG-TXL compounds described herein
  • the inventors also contemplate that other sterically similar compounds may be formulated to mimic the key portions of the water soluble polyamino acid structure.
  • Such compounds which may be termed peptidomimetics, may be used in the same manner as the peptides of the invention and hence are also functional equivalents.
  • peptide mimetics that mimic elements of protein secondary structure are described in Johnson et al. (1993).
  • the underlying rationale behind the use of peptide mimetics is that the peptide backbone of proteins, including polyamino acids, exists chiefly to orientate amino acid side chains in such a way as to facilitate molecular interactions, such as those of antibody and antigen.
  • a peptide mimetic is thus designed to permit molecular interactions similar to the natural molecule.
  • ⁇ -turn structure within a polypeptide can be predicted by computer-based algorithms, as discussed herein. Once the component amino acids of the turn are determined, mimetics can be constructed to achieve a similar spatial orientation of the essential elements of the amino acid side chains.
  • modified or unusual amino acids are also contemplated for use in the present invention.
  • a table of exemplary, but not limiting, modified or unusual amino acids is provided herein below. TABLE 1 Modified and Unusual Amino Acids Abbr.
  • Toxicity studies, pharmacokinetics and tissue distribution of DTPA-paclitaxel have shown that in mice the LD 50 (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 constraints 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 maximum tolerated doses (MTD) in a clinical study for use in human subjects.
  • MTD maximum tolerated doses
  • 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 proliferation
  • 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.
  • 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 microtubule 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 water soluble polyamino acids conjugated paclitaxel of the present invention are contemplated to be useful in the treatment of rheumatoid arthritis.
  • the polymer conjugated formulation disclosed herein would also offer the advantages of controlled 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.
  • 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.
  • the present example concerns a first study involving the conjugation of paclitaxel to a water-soluble polymer, poly (l-glutamic acid) (PG) and the efficacy of the preparation against a variety of tumors in mice and rats.
  • PG poly (l-glutamic acid)
  • the potential of water-soluble polymers used as drug carriers is well established (Kopecek, 1990; Maeda and Matsumura, 1989).
  • 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), Ara-C (Kato et al., 1984) and melphalan (Morimoto et al., 1984) have been conjugated to PG.
  • poly-aspartic acid may be conjugated to anti-tumor drugs using the reaction scheme described herein for PG-TXL.
  • PG 75 mg, repeating unit FW 170, 0.44 mmol
  • DMF dry N,N-dimethylformamide
  • DCC dicyclohexylcarbodiimide
  • DMAP trace amount of dimethylaminopyridine
  • Paclitaxel was supplied by Hande Tech (Houston, Tex.), and the purity was 99% and higher as confirmed by HPLC assay.
  • the sodium salt of PG-TXL conjugate was obtained by dissolving the product in 1.0 M NaHCO 3 .
  • the aqueous solution of PG-TXL was dialyzed against distilled water (MWCO 10,000) to remove low molecular weight contaminants and excess NaHCO 3 salt. Lyophilization of the dialysate yielded 98 mg of product as a white powder.
  • the paclitaxel content in this polymeric conjugate as determined by UV was 20-22% (w/w). Yield: 98% (conversion to polymer bound paclitaxel, UV). Solubility in water>20 mg paclitaxel/ml.
  • a similar method can be used to synthesize PG-TXL with higher paclitaxel content (up to 35%) by simply increasing the ratio of paclitaxel to PG used.
  • Resonances at 7.75 to 7.36 ppm are attributable to aromatic components of paclitaxel resonances at 6.38 ppm (C 120 -H), 5.97 ppm (C 13 -H), 5.63 ppm (C 2 ′-H, d), 5.55-5.36 ppm (C 3 ′-H and C 2 -H, m), 5.10 ppm (C 5 -H), 4.39 ppm (C 7 -H), 4.10 (C 20 -H), 1.97 ppm (OCOCH 3 ), and 1.18-1.20 ppm (C CH 3 ) are tentatively assigned to aliphatic components of paclitaxel. Other resonances were obscured by the resonances of PG.
  • PG resonances at 4.27 ppm (H- ⁇ ), 2.21 ppm (H- ⁇ ), and 2.04 ppm (H- ⁇ ) are in accordance with pure PG spectrum. Although a peak at 5.63 ppm could be tentatively assigned to the C-2′ proton of the C-2′ ester, the C-2′ proton of unsubstituted paclitaxel at 4.78 ppm was also present, suggesting that the resulting conjugate may contain paclitaxel substitutions at both the C-2′ and C-7 positions.
  • a 100 mg/ml solution of the conjugate produces a clear, viscous, yet flowable liquid. This procedure consistently produces PG-TXL conjugate containing 20% of paclitaxel by weight, i.e., approximately 7 paclitaxel molecules are bound to each polymer chain.
  • the relative molecular weight of PG-TXL 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 ultraviolet (UV) detection set at 270 nm.
  • UV ultraviolet
  • PG-TXL sodium salt a TSK-gel column suitable for analysis of water-soluble polymer was used, and the system was eluted with 0.2 mM PBS (pH 6.8) at 1.0 ml/min.
  • the tumor growth delay induced by PG-TXL was measured in mammary ovarian carcinoma (OCA-I) implanted in C3Hf/Kam mice. All tumors were syngeneic to this strain. Solitary tumors were produced in the muscle of the right thigh of female C3H/Kam mice (25-30 g) by injecting 5 ⁇ 10 5 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 ⁇ 10 5 viable 13762F tumor cells in 0.1 ml PBS. Treatments were initiated when the tumors in mice had grown to 500 mm 3 (10 mm in
  • PG-TXL was disolved in saline (10 mg equivalent paclitaxel/ml), and paclitaxel was dissolved in Cremophor EL(g vehicle (6 mg/ml). Data are presented as mean ⁇ standard deviation of tumor volumes. In control studies, saline (0.6 ml), Cremophor vehicle [50/50 Cremophor/ethanol diluted with saline (1:4)], PG solution in saline, and paclitaxel plus PG were used.
  • the maximum tolerated dose (MTD) of PG-TXL and paclitaxel in normal female C3Hf/Kam mice was estimated to be 160 mg/kg and 80 mg/kg respectively.
  • a single dose of PG-TXL in saline or paclitaxel in Cremophor EL vehicle was given in doses varying from 40 to 160 mg equiv. Paclitaxel/kg body weight.
  • Tumor growth was determined daily (FIGS. 7A, 7B, 7 C, 7 D and 7 E) by measuring three orthogonal tumor diameters. Tumor volume was calculated according to formula (A ⁇ B ⁇ 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 3 in mice minus the time in days for tumors treated with saline control to grow from 500 to 2,000 mm 3 .
  • mice When the tumor size reached 2000 mm 3 , the tumor growth delay was calculated; the mice were sacrificed when tumors were approximately 2500 mm 3 .
  • Table 2 summarizes acute toxicity of PG paclitaxel in rats in comparison with paclitaxel/Cremophor.
  • Table 3 summarizes the data concerning the effect of PG-TXL against MCa-4, FSa-II and HCa-I tumors in mice. The data are also summarized in FIG. 7A-FIG. 7E.
  • PG-TXL is most effective against MCa-4 and OCA-1 tumors.
  • PG-TXL is more effective than paclitaxel on equivalent mg paclitaxel basis in the case of MCa-4, HCa-I, and on OCA-1 tumors, and is remarkably potent at its maximum tolerated dose (MTD).
  • MTD maximum tolerated dose
  • a single dose of PG-TXL in saline or paclitaxel in a Cremophor EL® vehicle was given in doses varying from 20 to 60 mg equivalent paclitaxel/kg body weight.
  • saline, the Cremophor EL® vehicle [50/50 Cremophor/ethanol diluted with saline (1:4)]
  • PG solution in saline and paclitaxel plus PG were used.
  • complete tumor eradication at the MTD of PG-TXL 60 mg equivalent paclitaxel/kg
  • PG-TXL given at a lower dose of 40 mg equivalent paclitaxel/kg also resulted in complete tumor regression (FIG. 7B).
  • the MTD of paclitaxel in Cremophor EL® was less than 20 mg/kg.
  • Paclitaxel at this dose caused a tumor growth delay (Tumor growth delay is defined as the time in days for tumors treated with the test drugs to grow from 2,000 mm 3 to 10,000 mm 3 minus the time in days for tumors treated with saline control to grow from 2,000 mm 3 to 10,000 mm 3 .) of only 5 days, whereas the same equivalent paclitaxel dose of PG-TXL resulted in a tumor growth delay of 23 days (FIG. 7B).
  • mice were injected with 2 ⁇ 10 6 MDA-435-Lung2 cells (a variant of the MDA-MB-435 human breast cancer cell line) into the mammary fatpad. When the tumors reached 5 mm mean diameter, (27 days after tumor injection), mice were treated with an i.v. injection of PG-TXL or the various controls (see Table 4). Tumor measurements were taken weekly. Tumors that reached 1.5 cm were removed surgically. All mice were killed at 120 days, and remaining tumors removed and weighed. Mice were examined for metastases, and lungs processed for histology, with single sections of the organs scored for the presence of micrometastases. TABLE 4 No.
  • Tumor Mean tumor tumors Lung Treatment take a wt (g) b regressed c metastases d PBS 5/6 1.3 ⁇ 0.24 — 4/5 (80%) Cremophor 9/9 1.26 ⁇ 0.67 — 4/8 (50%) PGA 10/10 1.13 ⁇ 0.7 — 4/7 (57%) Taxol TM/Cremophor 10/10 1.31 ⁇ 0.69 — 3/7 (42%) 60 mg/kg PG-TXL 60 mg/kg 10/10 1.23 ⁇ 0.38 2/10 5/8 (62.5%) PG-TXL 120 mg/kg 9/10 0.925 ⁇ 0.12 4/8 1/4 (25%) #equivalent of PG-TXL.
  • mice were injected with 2 ⁇ 10 6 ; MDA-435-Lung 2 cells (a variant of the MDA-MB-435 human breast cancer cell line) into the mammary fatpad.
  • MDA-435-Lung 2 cells a variant of the MDA-MB-435 human breast cancer cell line
  • the treatments were started, and repeated at 14 day intervals (day 24, 38, 52) for a total of three injections. Tumor measurements were taken weekly. The mice were killed on day 105 after tumor cell injection, and the tumor weights and incidence of metastasis recorded. The lungs were processed for histology, and single sections scored for the presence of micrometastases. The results are shown in Table 5.
  • mice were injected i.p. with the human ovarian cancer cell line, SKOV3ip1.
  • the mice were injected i.v. with the PG-paclitaxel (PG-TXL), at concentrations equivalent to 120 mg/kg or 160 mg/kg of paclitaxel. Initially the plan was to repeat these injections at 7-day intervals, but a single injection of the 160 mg/kg dose killed 5 of the 10 mice. Only the 120 mg/kg group received three injections. The study was terminated on day 98, and any surviving mice killed. The results are shown in FIG. 14, and in Table 6.
  • the PG-TXL 120 mg/kg significantly extended the survival of the mice with intraperitoneal SKOV3ipl, (a human ovarian cancer cell line which overexpresses HER2/neu), compared with mice injected with PG alone. Multiple doses and/or increasing the dose of conjugate may significantly reduce the tumor incidence in addition to extending survival.
  • SKOV3ipl a human ovarian cancer cell line which overexpresses HER2/neu
  • the growth curves show that although breast cancer growth is checked by paclitaxel, especially with the higher dose conjugated with PG, tumor size continues to increase about a month after the therapy. A second (or third) round of therapy may have caused the tumor growth to plateau, or give more tumor regressions.
  • the growth curves do not include the tumors that regressed—as shown in Table 4, the tumors shrank/disappeared in 50% of the mice treated with the highest dose of PG-TXL, and of the 4 animals with progressively growing tumors at the end of the study, only one had micrometastases in the lungs. So the treatment that reduced growth of the primary tumors also reduced the incidence of metastasis.
  • paclitaxel 100 mg, 0.1 17 mmol
  • dry DMF 2.2 ml
  • DTPA A diethylenetriaminepentaacetic acid anhydride
  • 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).
  • Paclitaxel had an R f value of 0.34.
  • 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).
  • 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 ⁇ 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.
  • 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.
  • FIG. 2 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.
  • mice Female C3Hf/Kam mice were inoculated with mammary carcinoma (MCa-4) in the muscles of the right thigh (5 ⁇ 10 5 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. 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).
  • HPLC HPLC was used to analyze the reaction mixture and purity of 111 In-DTPA-paclitaxel.
  • the system consisted of a LDC binary pump, a 100 ⁇ 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 NaI crystal detector and a Spectra-Physics UV/Vis detector. As evidenced by HPLC analysis, purification by Sep-Pak cartridge removed most of the 111 In-DTPA, which had a retention time of 2.7 min.
  • the 111 In-DTPA was probably derived from traces of DTPA contaminant in the DTPA-paclitaxel.
  • a radio-chromatogram of 111 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.
  • mice Female C3Hf/Kam mice were inoculated with mammary carcinoma (MCa-4) in the muscles of the right thigh (5 ⁇ 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 111 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 h after injection.
  • MCa-4 mammary carcinoma
  • FIG. 5 shows gamma-scintigraphs of animals injected with 111 In-DTPA and 111 In-DTPA-paclitaxel.
  • 111 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.
  • 111 In-DTPA-paclitaxel exhibited a pharmacological profile resembling that of paclitaxel (Eiseman et al., 1994). Radioactivity in the brain was negligible.
  • PEG-paclitaxel was synthesized by an N-ethoxycarbonyl-2-ethoxy-1,2-dihydroquinoline (EEDQ) mediated coupling reaction.
  • EEDQ N-ethoxycarbonyl-2-ethoxy-1,2-dihydroquinoline
  • 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).
  • GPC analysis gel permeation chromatography
  • 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.
  • melanoma cells were seeded in 24-well plates at a concentration of 2.5 ⁇ 10 4 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 h in a 97% humidified atmosphere of 5.5% CO,. The medium was then replaced with fresh medium containing paclitaxel or its derivatives in concentrations ranging from 5 ⁇ 10 ⁇ 9 M to 75 ⁇ 10 ⁇ 9 M. After 40 h, the cells were released by trypsinization and counted in a Coulter counter.
  • DME Dulbecco's modified minimal essential medium
  • F12 medium containing 10% bovine calf serum
  • 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 ⁇ 10 ⁇ 9 M to 75 ⁇ 10 ⁇ 9 M also did not effect cell proliferation.
  • MCa-4 cells (5 ⁇ 10 5 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 min of injection.
  • PEG-paclitaxel was dissolved in saline (6 mg equiv. paclitaxel/ml) and filtered through a sterile filter (Millipore, 4.5 elm). 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 studies. 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 ( ⁇ >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).
  • the objective of this study was to compare PG-TXL and paclitaxel pharmacological properties in their ability to promote in vitro assembly of tubulin, to inhibit cell growth against rat mammary tumor cell line 13762F and human breast tumor cell lines, to induce p53 protein, and to rescue a paclitaxel-dependent mutant cell line.
  • Paclitaxel's release from PG-TXL in vivo was measured to determine if PG-TXL's mechanism of action can be attributed to free pacitaxel.
  • paclitaxel PG-TXL
  • PG-TXL PG-TXL
  • aged PG-TXL were compared for their relative ability to promote in vitro assembly of purified bovine brain tubulin.
  • the tubulin assembly reaction was performed at 32° C.
  • PEM buffer 80 mM PIPES buffer, 1 mM EGTA, 1 mM MgCl 2 , pH 6.9 at a tubulin (bovine brain, Cytoskeleton Inc., Boulder, Colo.) concentration of 1 mg/ml (10 ⁇ M) in the presence or absence of drugs (1.0 ⁇ M equivalent paclitaxel) and 1.0 mM guanosine 5′-triphosphate (GTP).
  • Tubulin polymerization was followed by measuring the absorbance of the solution at 340 nm over time.
  • PG-TXL and paclitaxel were compared for their ability to inhibit cell growth against the established rat mammary tumor cell line 13762F.
  • the effect of PG-TXL on cell growth was examined by a plating efficiency assay.
  • Rat 13762F cells were seeded (200 cells) into 60 mm dishes containing drug concentrations varying from 0 to 200 nM in growth medium (cc modified minimum essential medium [ ⁇ -MEM] containing 5% fetal bovine serum, 50 U/ml of penicillin, and 50 ⁇ g/ml of streptomycin).
  • IC 50 50% inhibition of colony formation
  • PG-TXL Since the “aged” solution is more potent than freshly dissolved PG-TXL, the in vitro degradation of PG-TXL or release of active drug appears to be important for PG-TXL to exert this biological activity. However, even after “aging,” PG-TXL is still 25 times less potent than paclitaxel.
  • Tax 18 a CHO cell line selected for resistance to paclitaxel, is a well characterized mutant that has been found to require the continuous presence of paclitaxel for cell division.
  • a functional mitotic spindle apparatus is unable to form (Cabral et al., 1983).
  • the mitosis phase of the cell cycle is prolonged with subsequent failure to segregate chromosomes and to divide into daughter cells. Nonetheless, the cells continue to progress through the cell cycle and replicate their DNA resulting in the formation of large polyploid cells that eventually die due to genomic instability (Cabras and Barlow, 1991).
  • paclitaxel-dependent CHO mutant Tax-18 cells were carried out on 24-well tissue culture dishes. Approximately 100 cells were added to wells containing growth medium and equivalent concentrations of paclitaxel varying from 0 to 1.0 ⁇ M. After 6 days of incubation at 37° C., the medium was removed and the cells were stained with methylene blue.
  • mice normal female C3Hf/Kam mice (25-30 g) were injected with a dose of 20 mg equivalent [ 3 H]paclitaxel or PG-[ 3 H]paclitaxel intravenously into the tail vein. Each mouse received 6 ⁇ Ci of radiolabeled drug. [ 3 H]paclitaxel was dissolved in Cremophor EL® vehicle whereas PG-[ 3 H]paclitaxel was dissolved in saline. Volume injected into each mice was between 0.2 to 0.3 ml. At 0, 5, 15, 30 min, and 1, 2, 4, 8, 16, 24, 48 h postinjection, animals were sacrificed and blood samples were collected (4-5 mice per time point).
  • mice bearing OCA-1 tumors were prepared as described previously. When the tumor reached 500 mm 3 , animals were injected with a dose of 20 mg equivalent paclitaxel/kg of [ 3 H]paclitaxel or PG-[ 3 H]TXL into the tail vein. Animals were killed at 2, 5, 9, 24, 48, and 144 h postinjection. Tumors were removed, weighed, and homogenized with 3 volume of PBS (w/v). Percent of injected dose per gram tissue is calculated based on total radioactivity associated with the tumor, which was determined by counting prepared tissue homogenate aliquots.
  • tissue homogenate was mixed with tissue solubilizer, followed by addition of scintillation solvent, and counted for total radioactivity. The counting efficiency was verified by the method of standard addition.
  • aliquots of tissue homogenates were extracted with ethyl acetate and analyzed for free paclitaxel by HPLC.
  • the HPLC system consisted of a 150 ⁇ 3.9 mm Nova-Pak column (Waters, Milford, MA), a liquid chromatography pump (Waters model 510), a UV/Vis detector set at 228 nm (Waters model 486), a flow scintillation analyzer (Packard model 500TR, Downers Grove, Ill.), and a Packard radiomatic software for data analysis.
  • the uptake of total drugs in OCA-1 tumor was expressed as a percentage of the administered dose per gram of tissue and the association of radioactivity within OCA-1 tumor as free paclitaxel was expressed as dpm per gram tissue.
  • paclitaxel/kg body weight produced a tumor growth delay of 21 days vs. saline treated controls.
  • Poly(d-glutamic acid)-paclitaxel was as effective as poly(l-glutamic acid)-paclitaxel.
  • paclitaxel conjugated with poly(l-aspartic acid) was completely inactive against OCA-1 tumor.
  • the antitumor activity of polymer-paclitaxel conjugates of different molecular weight (1K, 13K, and 36K) was compared. Conjugates of lower molecular weight were significantly less effective than conjugate of higher molecular weight. The higher molecular weights above 50,000 was too viscous.
  • OCA-1 tumor bearing mice were prepared as previously described. When tumor volume reached 500 mm 3 , animals were injected with either paclitaxel (80 mg/kg) or PG-TXL (160 mg equivalent paclitaxel/kg). At different times ranging from 0 to 144 h after treatment, tumors were histologically analyzed to quantify mitotic and apoptotic activity according to Milas et al. (1995). The mice were killed by cervical dislocation and the tumors were immediately excised and placed in neutral-buffered formalin.
  • the tissues were then processed and stained with hematoxylin and eosin. Both mitosis and apoptosis were scored in coded slides by microscopic examination at 400 ⁇ magnification. Five fields of nonnecrotic areas were randomly selected in each histological specimen, and in each field the number of apoptotic nuclei and cells in mitosis were recorded as numbers per 100 nuclei and were expressed as a percentage. The values were based on scoring 1500 nuclei obtained from 3 mice per time point.
  • tumors from PG-TXL-treated mice developed extensive edema and necrosis, and only a small rim of viable tumor cells remained.
  • the residual tumor clumps as compared to controls were comprised of cells that were larger, more pleomorphic, and that displayed less mitotic activity.
  • the content of CPT in the polymer conjugate was determined by fluorescence spectrometer (Perkin-Elmer Model MPF-44A) using emission wavelength of 430 nm and excitation wavelength of 370 nm. Content: 2% to 5% (w/w), solubility: >200 mg conjugate/ml.
  • All accessible amine functional groups of poly-lysine (MW>30,000, Sigma) will be converted to carboxylic acid functional groups by reacting poly-lysine with succinic anhydride, glutaric anhydride, or DTPA. The remaining unreacted NH2 group in poly-lysine will be blocked by reacting the modified polymer with acetic anhydride.
  • TXL, docetaxel, other taxiods, etopside, teniposide, camptothecin, epothilone or other anti-tumor drugs will be conjugated to the resulting polymer according to previously described procedures for the synthesis of PG-TXL.
  • Polyamino acid copolymers containing glutamic acid may be synthesized by the copolymerization of N-carboxyanhydrides (NCAs) of corresponding amino acid with gamma-benzyl-L-glutamate NCA.
  • NCAs N-carboxyanhydrides
  • the resulting benzyl glutamate-containing copolymer will be converted to glutamic acid-containing copolymer by removing the benzyl protecting group (FIG. 15).
  • TXL docetaxel
  • other taxiods etopside, teniposide, camptothecin, epothilone or other anti-tumor drugs
  • PG-TXL docetaxel
  • PG-CPT PG-CPT
  • Poly-L-glutamic acid-Paclitaxel is a conjugate of poly-L-glutamic acid and paclitaxel.
  • This compound is water soluble and based on early animal studies it appears that it can be administered as a short, that is several minute, intravenous injection. Based on the in vitro and early animal work, it appears that this compound is at least as active against cancer as the monomeric paclitaxel in Cremophor and may have fewer side effects. Based on these observations, this drug will be studied in humans. The study will first require formulation of this compound in a solvent which is commonly used for intravenous infusion.
  • Phase I study of this drug will be performed using the starting dose defined in animal studies.
  • the drug will be injected into the vein by a syringe over several min or alternatively it may be infused as a short infusion, up to approximately 10 to 15 min.
  • the volume of the solvent will be from 10 ml to approximately 100 ml depending on which of the two intravenous injection approaches are used.
  • the drug will be administered every 3 wk. This schedule is based on the early animal studies and on the schema used with paclitaxel in Cremophor. Three patients will be started on the lowest dose level as defined by the animal studies and will be treated with an injection of PG-TXL.
  • Blood tests will be performed at baseline and weekly to evaluate blood counts; tests of liver function and renal function will be performed every 3 wk. It is expected that the counts and physiological parameters will recover sufficiently from the PG-TXL to resume the next cycle of treatment 3 wk after the previous one. If this is the case then the treatment will be repeated every 3 wk. If the first cohort of three patients tolerates the drug for 3 wk then these patients will be allowed to have the dose increased by a predetermined schema that is usually used in the Phase I studies. Once three patients have tolerated the first cycle, the next cohort of 3 patients will be started on the next higher dose level. This process of increasing the dose level will continue until at least 2 out of 3 patients at a dose level have side effects which are so severe that they prohibit continuing administration of the drug.
  • the dose level just prior to the excessively toxic one will be considered the level of drug to be administered in subsequent studies.
  • Six to ten patients shall be treated on the dose level which will be recommended for Phase II studies to confirm its tolerability. Once the appropriate dose has been defined and acute toxic side effects of the drug evaluated, Phase II studies will be initiated.
  • Phase II studies of PG-TXL will be performed in several tumor types. Each study will be designed in a usual standard Phase II manner following either Gahan's or Simon's design. In brief, approximately 14 patients of a given tumor type will be treated initially, if there is no evidence of anti-cancer activity in that tumor type then further studies of PG-TXL in that tumor type will be aborted. However, if at least one patient has clinical benefit, defined as at least 50% decrease in the sum of products of perpendicular cross-sectional diameters of the tumors, then the number of patients with that tumor type treated with PG-TXL will be increased to 30.
  • the tumor types of special interest for PG-TXL will be the ones which have shown good response to paclitaxel and docetaxel. This will include ovarian cancer, breast cancer, and lung cancer. Studies comparing poly-glutamic acid-paclitaxel to paclitaxel in tumors showing response to PG-TXL will be performed. Such studies are called Phase III studies.
  • PG-TXL may be cost effective relative to paclitaxel monotherapy.
  • the infusion going to be shorter it is expected that in view of the absence of Cremophor fewer side effects will be experienced by the patients and therefore the premedication regiment including steroids and intravenous H2 and H1 blockers may no longer be necessary. All of these factors will result in a reduction in the cost of the treatment.
  • Phase II studies in various tumor types may take another 6 to 9 months.
  • the inventors will have a good idea of the efficacy of this drug and targeted Phase III studies may be designed and initiated. It is also possible that the Phase II studies will show enough clinical activity that abbreviated Phase III studies or no Phase III studies would be necessary.
  • 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.

Abstract

Disclosed are water soluble compositions of paclitaxel and docetaxel formed by conjugating the paclitaxel or docetaxel to a water soluble polymer such as poly-glutamic acid, poly-aspartic acid or poly-lysine. Also disclosed are methods of using the compositions for treatment of tumors, auto-immune disorders such as rheumatoid arthritis. Other embodiments include the coating of implantable stents for prevention of restenosis.

Description

  • This application is a continuation-in-part of co-pending application U.S. Ser. No. 08/815,104, which is a continuation of U.S. Provisional Application No. 60/013,184.[0001]
  • 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 docetaxel (Taxotere), in particular making paclitaxel water soluble by conjugating the drug to water soluble moieties. [0002]
  • BACKGROUND OF THE INVENTION
  • Paclitaxel, an anti-microtubule agent extracted from the needles and bark of the Pacific yew tree, [0003] 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 [0004] 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). Paclitaxel (Taxol™) has shown significant activity in human cancers, including breast, ovarian, non-small cell lung, and head and neck cancers (Rowinsky and Donehower, 1995). It has also shown significant activity in patients with advanced breast cancer who had been treated with multiple chemotherapeutic agents (Foa et al., 1994). As with most chemotherapeutic agents, however, the maximum tolerated dose of paclitaxel is limited by toxicity. In humans, paclitaxel's major toxic effect at doses of 100-250 mg/m[0005] 2 is granulocytopenia (Holmes et al., 1995); symptomatic peripheral neuropathy is its principal nonhematologic toxicity (Rowinsky et al., 1993).
  • 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 vasodilatation, 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 particulate 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. [0006]
  • Prior attempts to obtain water soluble paclitaxel have included the preparation of prodrugs of paclitaxel by placing solubilizing moieties such as succinate, sulfonic acid, amino acids, and phosphate derivatives at the 2′-hydroxyl group or at the 7-hydroxyl position (Deutsch et al., 1989; Mathew et al., Zhao and Kingston, 1991, 1992; Nicolaou et al., 1993; Vyas et al., 1995, Rose et al., 1997). While some of these prodrugs possess adequate aqueous solubility, few have antitumor activity comparable to that of the parent drug (Deutsch et al., 1989; Mathew et al., 1992; Rose et al., 1997). Several of these derivatives are not suitable for i.v. injection because of their instability in aqueous solution at neutral pH. 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 reported to be unstable. Similar results were reported by Mathew et al. (1992). [0007]
  • Recently, Nicolaou et al. (1993) reported the synthesis and in vitro biological evaluation of a novel type of prodrug termed “protaxols”. These compounds possess greater aqueous solubility and are converted to paclitaxel as the active drug through an intramolecular hydrolysis mechanism. However, no in vivo data on the antitumor activity of protaxols are yet available. Greenwald et al. reported the synthesis of highly water-soluble 2′ and 7-polyethylene glycol esters of paclitaxel (Greenwald et al., 1994). Using the strategy of polymer linkage, others have developed water-soluble polyethylene glycol (PEG)-conjugated paclitaxel (Li et al., 1996; Greenwald et al., 1996). Although these conjugates have excellent water solubility, their therapeutic efficacies are not better than free paclitaxel. Moreover, PEG has only two reactive functional groups at each end of its polymer chain, which effectively limit the amount of paclitaxel that PEG could carry (U.S. Pat. No. 5,362,831). [0008]
  • Other 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. [0009]
  • 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 angioplasty and stenting. [0010]
  • 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/or antiangiogenic drug, such as paclitaxel, docetaxel, or other taxoid conjugated to a water soluble polymer such as a water soluble polyamino acid, or to a water soluble metal chelator. It is a further embodiment of the present invention that a composition comprising a conjugate of paclitaxel and poly-glutamic acid has surprising antitumor activity in animal models, and further that this composition is demonstrated herein to be a new species of taxane that has pharmaceutical properties different from that of paclitaxel. 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, docetaxel, or other taxoid 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 improved efficacy and controlled release so that tumors are shown herein to be eradicated in animal models after a single intravenous administration, as well as providing a novel taxane. Poly-(l-glutamic acid) conjugated paclitaxel is shown in the examples hereinbelow to have a novel drug activity, in addition to having improved the delivery to the tumor and providing a controlled release. [0011]
  • The methods described herein could also be used to make water soluble polymer conjugates of other therapeutic agents, contrast agents and drugs, including paclitaxel, tamoxifen, Taxotere, etopside, teniposide, fludarabine, doxorubicin, daunomycin, emodin, 5-fluorouracil, FUDR, estradiol, camptothecin, retinoids, verapamil, epothilones cyclosporin, and other taxoids. 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. [0012]
  • 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 paclitaxel, docetaxel, or other taxoid, or in preferred embodiments the poly-(l-glutamic) acid conjugated paclitaxel (PG-TXL), of the present invention may, in certain types of treatment, be combined with a platinum drug, an antitumor agent such as doxorubicin or daunorubicin, for example, or other drugs that are used in combination with Taxol™ or combined with external or internal irradiation. [0013]
  • 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). Tumors often lack a lymphatic vasculature to remove large molecules that leak into the tumor tissue (Maeda and Matsumura, 1989). 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. The water soluble polymers, such as, in preferred embodiments PG-TXL, may have pharmacological properties different from non-conjugated drugs (i.e. paclitaxel). Additionally, polymer-drug conjugates may act as drug depots for sustained release, producing prolonged drug exposure to tumor cells. Finally, water soluble polymers (e.g., water soluble polyamino acids) 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 known by the present inventors to be currently undergoing clinical evaluation, including SMANCS in Japan and HPMA-Dox in the United Kingdom (Maeda, 1991; Kopecek and Kopeckova, 1993). [0014]
  • 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 a preferred taxane is a chemical of the general chemical formula, C[0015] 47H5, NO14, including [2aR-[2aα,4β,4αβ,6β,9α(αR*,βS*), 11α, 12α, 12aα, 12βα,]]-β-(Benzoylamino)-α-hydroxybenzene propanoic 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-7,11-methano-1H-cyclodeca[3,4]benz-[1,2-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 or taxane 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 metals. Preferred radionuclides include, but are not limited to [0016] 67Ga, 68Ga, 111In, 99mTc, 90Y, 114mSn and 193mPt.
  • Preferred water soluble chelators to be used in the practice of the present invention include, but are not limited to, diethylenetriaminepentaacetic acid (DTPA), ethylenediaminetetraacetic acid (EDTA), 1,4,7,1 0-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 1-(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 [0017] 111In-DTPA-paclitaxel, and Na-DTPA-paclitaxel.
  • In certain embodiments of the present invention, the paclitaxel, docetaxel, or other taxoid 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, docetaxel, or other taxoid. Poly-glutamic acid (PG) is one polymer that offers several advantages in the present invention. First, it contains a large number of side chain carboxyl functional groups for drug attachment. Second, PG can be readily degraded by lysosomal enzymes to its nontoxic basic component, 1-glutamic acid, d-glutamic acid and di-glutamic acid. [0018]
  • Finally, sodium glutamate has been reported to prevent manifestations of neuropathy induced by paclitaxel, thus enabling higher doses of paclitaxel to be tolerated (Boyle et al., 1996). Preferred polymers include, but are not limited to poly(l-glutamic acid), poly(d-glutamic acid), poly(dl-glutamic acid), poly(l-aspartic acid), poly(d-aspartic acid), poly(dl-aspartic acid), poly(l-lysine), poly(d-lysine), poly(dl-lysine), copolymers of the above listed polyamino acids with polyethylene glycol, polycaprolactone, polyglycolic acid and polylactic acid, as well as poly(2-hydroxyethyl 1-glutamine), chitosan, carboxymethyl dextran, hyaluronic acid, human serum albumin and alginic acid, with poly-glutamic acids being particularly preferred. At the lower end of molecular weight, the polymers of the present invention preferably have a molecular weight of about 1,000, about 2,000, about 3,000, about 4,000, about 5,000, about 6,000, about 7,000, about 8,000, about 9,000, about 10,000, about 11,000, about 12,000, about 13,000, about 14,000, about 15,000, about 16,000, about 17,000, about 18,000, about 19,000, about 20,000, about 21,000, about 22,000, about 23,000, about 24,000, about 25,000, about 26,000, about 27,000, about 28,000, about 29,000, about 30,000, about 31,000, about 32,000, about 33,000, about 34,000, about 35,000, about 36,000, about 37,000, about 38,000, about 39,000, about 40,000, about 41,000, about 42,000, about 43,000, about 44,000, about 45,000, about 46,000, about 47,000, about 48,000, about 49,000, to about 50,000 kd. At the higher end of molecular weight, the polymers of the present invention preferably have a molecular weight of about 51,000, about 52,000, about 53,000, about 54,000, about 55,000, about 56,000, about 57,000, about 58,000, about 59,000, about 60,000, about 61,000, about 62,000, about 63,000, about 64,000, about 65,000, about 66,000, about 67,000, about 68,000, about 69,000, about 70,000, about 71,000, about 72,000, about 73,000, about 74,000, about 75,000, about 76,000, about 77,000, about 78,000, about 79,000, about 80,000, about 81,000, about 82,000, about 83,000, about 84,000, about 85,000, about 86,000, about 87,000, about 88,000, about 89,000, about 90,000, about 91,000, about 92,000, about 93,000, about 94,000, about 95,000, about 96,000, about 97,000, about 98,000, about 99,000, to about 100,000 kd. Within these ranges, the ranges of molecular weights for the polymers are preferably of about 5,000 to about 100,000 kd, with about 20,000 to about 80,000 being preferred, or even about 25,000 to about 50,000 being more preferred. [0019]
  • It is a further aspect of the invention that a composition of the invention such as PG-TXL may also be conjugated to a second lipophilic or poorly soluble antitumor agent such as camptothecin, epothilone, cisplatin, melphalan, Taxotere, etoposide, teniposide, fludarabine, verapamil, or cyclosporin, for example, or even to water soluble agents such as 5 fluorouracil (5 FU) or fluorodeoxyuridine (FUDR), doxorubicin or daunomycin. [0020]
  • 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, docetaxel, or other taxoid conjugated to a high molecular weight water soluble polymer or to a chelator. The pharmaceutical composition may include polyethylene glycol, poly-glutamic acids, poly-aspartic acids, poly-lysine, 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. [0021]
  • In certain embodiments, the present invention may be described as a method of determining the uptake of a chemotherapeutic drug such as paclitaxel, docetaxel, or other taxoid 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. [0022]
  • 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, docetaxel, or other taxoid 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, docetaxel, or other taxoid conjugated to a water soluble polyamino acids, including but not limited to poly (l-aspartic acid), poly (d-aspartic acid), or poly (dl-aspartic acid), poly (l-lysine acid), poly (d-lysine acid), or poly (dl-lysine acid), and more preferably to poly (l-glutamic acid), poly (d-glutamic acid), or poly (dl-glutamic 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, head and neck cancer. The compositions of the invention may also be used against gastric cancer, prostate cancer, colon cancer, leukemia, or Kaposi's Sarcoma. As used herein the term “treating” cancer is understood as meaning any medical management of a subject having a tumor. The term would encompass any inhibition of tumor growth or metastasis, or any attempt to inhibit, slow or abrogate tumor growth or metastasis. The method includes killing a cancer cell by non-apoptotic as well as apoptotic mechanisms of cell death. 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, by methods that include but are not limited to bolus injection or infusion, as well as intraarterial, intravenous, intraperitoneal, or intratumoral administration of the drug. [0023]
  • 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. [0024]
  • 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. [0025]
  • 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 polymer, with polyamino acids being preferred and poly-glutamic acid being more preferred. 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 paclitaxel when administered in the standard Cremophor formulation (U.S. Pat. No. 5,583,153, incorporated herein by reference). As in the treatment of tumors, it is contemplated that the effectiveness of the water soluble taxoids or taxane of the present invention will not be diminished by the conjugation to a water soluble moiety. Therefore, the compositions of the present invention are expected to be as effective as paclitaxel against rheumatoid arthritis. Paclitaxel is an antiangiogenic agent. Rheumatoid arthritis creates a collection of newly formed vessels which erode the adjacent joints. It is also understood that the taxoid or taxane 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 other anti-cancer drugs, such as methotrexate. [0026]
  • 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, incorporated herein by reference). 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 stent 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-TXL 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-glutamic acid or other water soluble poly-amino acids. In the practice of the method, the subject may be a coronary bypass, vascular surgery, organ transplant or coronary or any other arterial angioplasty patient, for example, and the composition may be administered directly, intravenously, or even coated on a stent to be implanted at the sight of vascular trauma. [0027]
  • 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 poly-glutamic acids or water soluble polyamino 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 during or after balloon angioplasty and the coating is effective to inhibit restenosis. [0028]
  • In certain preferred embodiments, the invention may be described as a composition comprising poly-glutamic acids conjugated to the 2′ or 7 hydroxyl or both of paclitaxel, docetaxel, or other taxoids, or even a composition comprising water soluble polyamino acids conjugated to the 2′ or 7 hydroxyl or both of paclitaxel, docetaxel, or other taxoids. [0029]
  • As used herein, the terms “a poly-glutamic acid” or “poly-glutamic acids” include poly (l-glutamic acid), poly (d-glutamic acid) and poly (dl-glutamic acid), the terms “a poly-aspartic acid” or “poly-aspartic acids” include poly (l-aspartic acid), poly (d-aspartic acid), poly (dl-aspartic acid), the terms “a poly-lysine” or “poly-lysine” include poly (l-lysine), poly (d-lysine), poly (dl-lysine), and the terms “a water soluble polyamino acid”, “water soluble polyamino acids”, or “water soluble polymer of amino acids” include, but are not limited to, poly-glutamic acid, poly-aspartic acid, poly-lysine, and amino acid chains comprising mixtures of glutamic acid, aspartic acid, and/or lysine. In certain embodiments, the terms “a water soluble polyamino acid”, “water soluble polyamino acids”, or “water soluble polymer of amino acids” include amino acid chains comprising combinations of glutamic acid and/or aspartic acid and/or lysine, of either d and/or l isomer conformation. In certain prefered embodiments, such a “water soluble polyamino acid” contains one or more glutamic acid, aspartic acid, and/or lysine residues. Such “water soluble polyamino acids” may also comprise any natural, modified, or unusual amino acid described herein, as long as the majority of residues, i.e. greater than 50%, comprise glutamic acid and/or aspartic acid and/or lysine. In certain embodiments, a water soluble polymer of amino acids that contains more than one different type of amino acid residue is sometimes referred to herein as a “co-polymer”. [0030]
  • In certain embodiments, various substitutions of naturally occurring, unussual, or chemically modified amino acids may be made in the amino acid composition of the “water soluble polyamino acids”, and particularly in “poly-glutamic acids”, to produce a taxoid-polyamino acid conjugate of the present invention and still obtain molecules having like or otherwise desirable characteristics of solubility and/or therapeutic efficacy. A polyamino acid such as poly-glutamic acid, poly-aspartic acid, poly-lysine, or water soluble amino acids chain or polymer comprising a mixture of glutamic acid, aspartic acid, and/or lysine, may, at the lower end of the amino acid substitution range, have about 1, about 2, about 3, about 4, about 5, about 6, about 7, about 8, about 9, about 10, about 12, about 13, about 14, about 15, about 16, about 17, about 18, about 19, about 20, about 21, about 22, about 23, about 24, or about 25 or more glutamic acid, aspartic acid, or lysine, residues, respectively, substituted by any of the naturally occurring, modified, or unusual amino acids described herein. In other aspects of the invention, a polyamino acid such as poly-glutamic acid, poly-aspartic acid, poly-lysine, or a poly-amino acid chain comprising a mixture of some or all of these three amino acids may, at the lower end, have about 1%, about 2%, about 3%, about 4%, about 5%, about 6%, about 7%, about 8%, about 9%, about 10%, about 11%, about 12%, about 13%, about 14%, about 15%, about 16%, about 17%, about 18%, about 19%, about 20%, about 21%, about 22%, about 23%, about 24%, to about 25% or more glutamic acid, aspartic acid, or lysine residues, respectively, substituted by any of the naturally occurring, modified, or unusual amino acids described herein. [0031]
  • In further aspects of the invention, a polyamino acid such as poly-glutamic acid, poly-aspartic acid, or poly-lysine may, at the high end of the amino acid substitution range, have about 25%, about 26%, about 27%, about 28%, about 29%, about 30%, about 31%, about 32%, about 33%, about 34%, about 35%, about 36%, about 37%, about 38%, about 39%, about 40%, about 41%, about 42%, about 43%, about 44%, about 45%, about 46%, about 47%, about 48%, about 49%, to about 50% or so of the glutamic acid, aspartic acid, or lysine residues, respectively, substituted by any of the naturally occurring, modified, or unusual amino acids described herein, as long as the majority of residues comprise glutamic acid and/or aspartic acid and/or lysine. In amino acid substitution of the various water soluble amino acid polymers, residues with a hydrophilicity index of +1 or more are preferred. [0032]
  • In certain aspects of the invention, the amount of anti-tumor drug conjugated per water soluble polymer can vary. At the lower end, such a composition may comprise from about 1%, about 2%, about 3%, about 4%, about 5%, about 6%, about 7%, about 8%, about 9%, or about 10%, about 11%, about 12%, about 13%, about 14%, about 15%, about 16%, about 17%, about 18%, about 19%, about 20%, about 21% about 22%, about 23%, about 24%, to about 25% (w/w) antitumor drug relative to the mass of the conjugate. At the high end, such a composition may comprise from about 26%, about 27%, about 28%, about 29%, about 30%, about 31% about 32%, about 33%, about 34%, about 35%, about 36%, about 37%, about 38%, about 39%, to about 40% or more (w/w) antitumor drug relative to the mass of the conjugate. Preferred anti-tumor drugs include paclitaxel, docetaxel, or other taxoids, and preferred water soluble polymers include water soluble amino acid polymers. [0033]
  • In certain other aspects of the invention, the number of molecules of anti-tumor drug conjugated per molecule of water soluble polymer can vary. At the lower end, such a composition may comprise from about 1, about 2, about 3, about 4, about 5, about 6, about 7, about 8, about 9, about 10, about 11, about 12, about 13, about 14, about 15, about 16, about 17, about 18, about 19, to about 20 or more molecules of antitumor drug per molecule of water soluble polymer. At the higher end, such a composition may comprise from about 21, about 22, about 23, about 24, about 25, about 26, about 27, about 28, about 29, about 30, about 31, about 32, about 33, about 34, about 35, about 36, about 37, about 38, about 39, about 40, about 41, about 42, about 43, about 44, about 45, about 46, about 47, about 48, about 49, about 50, about 51, about 52, about 53, about 54, about 55, about 56, about 57, about 58, about 59, about 60 about 61, about 62, about 63, about 64, about 65, about 66, about 67, about 68, about 69, about 70, about 71, about 72, about 73, about 74, to about 75 or more molecules or more of antitumor drug per molecule of water soluble polymer. Preferred anti-tumor drugs include paclitaxel, docetaxel, or other taxoids, and preferred water soluble polymers include water soluble amino acid polymers. The preferred number of anti-tumor drug molecules conjugated per molecule of water soluble polymer is about 7 molecules of antitumor drug per molecule of water soluble polymer. [0034]
  • Water soluble amino acid polymers with various substitutions of residues conjugated to paclitaxel, docetaxel, or other taxoids are referred to as “biological functional equivalents”. These “biologically functional equivalents” are part of the definition of “water soluble polyamino acids” that are conjugated to taxoids, and may be identified by the assays described herein as well as any applicable assay that is known to those of skill in the art to measure improved aqueous solubility relative to the unconjugated taxoid or taxoids used to produce the particular water soluble amino acid polymer-taxoid composition. In other aspects of the invention, “biological functional equivalents” of water soluble amino acid-taxoid polymers may be further identified by improved anti-tumor cell activity, relative to the anti-tumor cell activity of the unconjugated water soluble amino acid polymer used to produce the particular water soluble amino acid polymer-taxoid composition by the assays described herein as well as any applicable assay that is known to those of skill in the art. The term “biologically functional equivalents” as used herein to describe this aspect of the invention is further described in the detailed description of the invention. [0035]
  • 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. Also as used herein, the term “a” is understood to include the meaning “one or more”. 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.[0036]
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1A. Chemical structure of paclitaxel, PEG-paclitaxel and DTPA-paclitaxel. [0037]
  • FIG. 1B. Chemical structure and reaction scheme for production of PG-TXL. [0038]
  • FIG. 2. Effect of paclitaxel, PEG-paclitaxel and DTPA-paclitaxel on proliferation of B16 melanoma cells. [0039]
  • FIG. 3. Antitumor effect of DTPA-paclitaxel on MCa-4 mammary tumors. [0040]
  • FIG. 4. Median time (days) to reach tumor diameter of 12 mm after treatment with paclitaxel, DTPA-paclitaxel and PEG-paclitaxel. [0041]
  • FIG. 5. Gamma-scintigraphs of mice bearing MCa-4 tumors following intravenous injection of [0042] 111In-DTPA-paclitaxel and 111In-DTPA. Arrow indicates the tumor.
  • FIG. 6. Hydrolytic degradation of PG-TXL as determined in PBS as a function of time at different pH levels. -⋄- represents percent paclitaxel released, -O- represents metabolite-1 produced. [0043]
  • FIG. 7A. Anti-tumor effect of PG-TXL against syngeneic OCA-I ovarian carcinoma tumor in female C3Hf/Kam mice. Drugs were injected intraveneously in a single dose. Data are presented as mean ± standard deviation of tumor volumes. a, Mice bearing OCA-l tumor were injected with -z,[0044] 900 -, PG control (800 mg/kg; n=9); -▴-, paclitaxel (80 mg/kg; n=7); -Δ-, paclitaxel (80 mg/kg) plus PG (800 mg/kg; n=5); --, PG-TXL (80 mg equiv. paclitaxel; n=6); or -
    Figure US20010034363A1-20011025-P00901
    -, PG-TXL (160 mg equiv. paclitaxel/kg; n=26).
  • FIG. 7B. Anti-tumor effect of PG-TXL against 13762F tumor in female rats. -[0045]
    Figure US20010034363A1-20011025-P00900
    - represents PG control (220 mg/kg; n=7), -▴- represents paclitaxel (20 mg/kg; n=5), -Δ- represents paclitaxel (40 mg/kg; n=7), -- represents PG-TXL (20 mg equivalent paclitaxel/kg; n=5), -
    Figure US20010034363A1-20011025-P00901
    - represents PG-TXL (40 mg or 60 mg equivalent paclitaxel/kg; n=9).
  • FIG. 7C. The antitumor effect of PG-TXL on mice bearing MCa-4 mammary carcinoma tumors. -□- 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-TXL (40 mg/kg), -⋄- represents response to PG-TXL (60 mg equiv. paclitaxel/kg), -[0046]
    Figure US20010034363A1-20011025-P00901
    - represents response to PG-TXL (120 mg/kg).
  • FIG. 7D. The antitumor effect of PG-TXL against soft-tissue sarcoma tumor (FSa-II) in mice. -□- 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); -[0047]
    Figure US20010034363A1-20011025-P00901
    - represents response to paclitaxel (80 mg/kg), -Δ- represents response to PG-TXL (160 mg equiv. paclitaxel/kg).
  • FIG. 7E. The antitumor effect of PG-TXL against syngeneic hepatocarcinoma tumor (HCa-I) in mice. -□- 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); -[0048]
    Figure US20010034363A1-20011025-P00901
    - represents response to PG-TXL (80 mg/kg), -Δ- represents response to PG-TXL (160 mg equiv. paclitaxel/kg).
  • FIG. 8. Release profile of paclitaxel from PEG-paclitaxel in phosphate buffer (pH 7.4). Release profiles of paclitaxel (-×-); from PEG-paclitaxel (-[0049]
    Figure US20010034363A1-20011025-P00901
    -) at pH 7.4 is shown.
  • FIG. 9. Antitumor effect of PEG-paclitaxel on MCa-4 mammary tumors. -□-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, -[0050]
    Figure US20010034363A1-20011025-P00901
    -represents a single dose of 40 mg/kg body weight of paclitaxel, -- represents PEG-paclitaxel at 40 mg equiv. paclitaxel/kg body weight.
  • FIG. 10. Tubulin polymerization assays performed at 32° C. in the presence of 1.0 mM GTP and 1.0 mg/ml of tubulin. -[0051]
    Figure US20010034363A1-20011025-P00900
    - represents paclitaxel (1.0 EM), -Δ- represents PG-TXL (10 μM equivalent paclitaxel) incubated in PBS (pH 7.4) at 37° C. for 3 days, -
    Figure US20010034363A1-20011025-P00901
    - represents freshly dissolved PG-TXL.
  • FIG. 11. Plasma clearance of radioactivity following an i.v. injection of PG-[[0052] 3H]paclitaxel and [3H]paclitaxel in C3Hf/Kam mice. -
    Figure US20010034363A1-20011025-P00900
    - represents PG-TXL radioactivity after injection of 6 μCi of radiolabeled PG-[3H]paclitaxel (20 mg equivalent paclitaxel/kg), -×- represents paclitaxel radioactivity after injection of 6 μCi of radiolabeled [3H]paclitaxel (20 mg equivalent paclitaxel/kg), -
    Figure US20010034363A1-20011025-P00901
    - represents “Paclitaxel” radioactivity released from injected PG-[3H]paclitaxel.
  • FIG. 12A. Time-dependent OCA-1 tumor content of radioactivity following injection of either PG-[[0053] 3H]paclitaxel and [3 H]paclitaxel into mice. Open bars represents PG-TXL radioactivity after injection of 6 μCi of radiolabeled PG-[3H]paclitaxel (20 mg equivalent paclitaxel/kg), filled bars represents paclitaxel radioactivity after injection of 6 μCi of radiolabeled [3H]paclitaxel (20 mg equivalent paclitaxel/kg).
  • FIG. 12B. Conversion of PG-[[0054] 3H]paclitaxel to [3H]paclitaxel within OCA-1 tumor. Total radioactivity measured after injection of 6 μCi of radiolabeled PG-[3H]paclitaxel is shown in open bars, “Paclitaxel” derived radioactivity released from injected PG-[3H]paclitaxel is shown in solid bars.
  • FIG. 13. Kinetics of apoptosis in OCA-1 tumors after a single i.v. dose of 160 mg equiv. paclitaxel/kg of PG-TXL (MTD) and 80 mg/kg paclitaxel (MTD). -□- represents the response to a single i.v. dose of PG-TXL (160 mg equiv. paclitaxel/kg MTD), -◯- represents response to paclitaxel (80 mg paclitaxel/kg MTD). [0055]
  • FIG. 14. Survival of nude mice with human ovarian cancer cells (SKOV3ip1) treated with PG-TXL. Five days after tumor injection, the mice were injected i.v. with the PG-paclitaxel (PG-TXL), or PG control. Injections of PG-TXL were administered every seven days (▾) in the 120 mg/kg group, but not the 160 mg/kg group. -▪- represents untreated mice. -▴- represents the response to multiple i.v. doses of PG. -▾- represents the response to an i.v. dose of PG-TXL (120 mg equiv. paclitaxel/kg), -♦- represents the response to an i.v. dose of PG-TXL (160 mg equiv. paclitaxel/kg). [0056]
  • FIG. 15. Chemical structure and reaction scheme for production of glutamic acid containing polyamino acids.[0057]
  • 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 (l-glutamic acid) conjugated paclitaxel (PG-TXL) 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 maximum tolerated dose (MTD) of the PG-TXL 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 wk after a single i.v. injection). In a parallel study, the antitumor activity of PG-TXL in rats with rat mammary adenocarcinoma (13762F) was examined. Again, complete tumor eradication at 40-60 mg equiv. paclitaxel/kg of PG-TXL was observed. These surprising results demonstrate that the polymer-drug conjugate, PG-TXL, successfully eradicates well established solid tumors in both mice and rats after a single intravenous injection. [0058]
  • In addition to the remarkable antitumor (breast, ovarian, etc.) data in syngeneic mice, good activity of PG-TXL against human breast cancer (MDA-435) and ovarian cancer (SKOV3ipl) in nude mice has recently been observed. Nude mice are special animals with incomplete immune systems in which human tumors can grow. [0059]
  • The data presented herein have led the present inventors to conclude that PG-TXL is a novel species of taxane that is pharmacologically distinct from previous paclitaxel or Taxol™ preparations. For example, the distribution of PG-TXL within plasma is distinct from free paclitaxel. While paclitaxel remains in the plasma of mice for an extremely short time, PG-TXL appears to remain for a much longer period. This is contemplated to offer a distinct advantage in that prolonged exposure of tumors to the drug may result in an enhanced response. The rate of conversion of PG-TXL to paclitaxel is slow, with less than 1% of the radioactivity from radiolabeled PG-TXL being recovered as radioactive paclitaxel within 48 h after injection of the paclitaxel-polymer complex. This finding suggests that the novel drug, PG-TXL, may produce death within tumor cells in a manner which is not simply due to the gradual release of paclitaxel itself. [0060]
  • Further evidence of the novelty of PG-TXL is that relatively high levels of radioactivity from radiolabeled PG-TXL appear in tumor tissue shortly after injection. However, only small amounts of radioactivity within tumor tissue are due to the release of free paclitaxel. Furthermore, the percent of radioactivity within tumor tissue due to paclitaxel itself does not appreciably increase with time suggesting again that PG-TXL is a minimal prodrug for the gradual release of paclitaxel. Uptake of PG-TXL versus paclitaxel has also been studied in a specialized human colon adenocarcinoma cell transport system. While radioactivity associated with radiolabeled PG-TXL readily gained entry into cells, only 10% of it was due to free paclitaxel. These data parallel that which was found in studies of tissue distribution and again suggest that there are several mechanisms or ways in which PG-TXL may lead to the death of cancer cells which are different from those for paclitaxel. [0061]
  • In another study, it was discovered that freshly prepared PG-TXL does not support the growth of paclitaxel-dependent cell lines suggesting that free paclitaxel is only slowly released from the polymer-paclitaxel complex and that the polymer-paclitaxel complex itself is not behaving pharmacologically as “Taxol™”. Aging will promote the degradation of PG-TXL and does increase the relative ability of the resulting material to support the growth of paclitaxel-dependent cells, but to a lesser extent than compared to free paclitaxel. [0062]
  • Recent analyses of tumor tissues from mice treated with paclitaxel suggests that, as expected, this drug results in the formation of many apoptotic bodies within the tumor itself. Apoptosis is a mechanism in which cells commit self-induced death or programmed cell death, a natural process used by an organism in wound healing and tissue remodeling. Tumors from mice treated with PG-TXL had far fewer apoptotic bodies compared to free paclitaxel but had an increased incidence of tumor necrosis and edema suggesting that paclitaxel and PG-TXL may result in tumor cell death by two distinctly different pathways. [0063]
  • These studies, and those described in the specific examples, demonstrate that PG-TXL is a new taxane which is not only extremely active against breast and ovarian cancers, and appears to have limited side affects. It is now clear that the polymer conjugation of paclitaxel results in a compound (PG-TXL) that has novel and greater over-all antitumor activity. [0064]
  • Another aspect of the present invention is the inclusion of molecules in the polymeric composition that are effective to target the therapeutic composition to a disease or tumor site or to a particular organ or tissue. Many of such targeting molecules are known in the art and may be conjugated to the water soluble anti-tumor compositions of the present invention. Examples of such molecules or agents would include, but not be limited to antibodies such as anti-tumor antibodies; anti-cell receptor antibodies; tissue specific antibodies; hormonal agents such as octreotide, estradiol and tamoxifen; growth factors; cell surface receptor ligands; enzymes; hypoxic agents such as misonidazole and erythronitroimidazole; and antiangiogenic agents. [0065]
  • Another composition of the present invention is DTPA-paclitaxel, 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, [0066] 111Indium 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 that also have different antitumor properties than unmodified paclitaxel. 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 predict the efficacy of an anti-cancer treatment. This information may be helpful in guiding the practitioner in the selection of patients to undergo chelator-paclitaxel therapy. [0067]
  • The paclitaxel may be rendered water-soluble in many ways: i.e. 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., [0068] 111In, 90Y, 166Ho, 68Ga, 99mTc) for nuclear imaging and/or for radiotherapy studies. The structures of paclitaxel, polyethylene glycol-paclitaxel (PEG-paclitaxel), poly-glutamic acid-paclitaxel conjugate (PG-TXL) 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 [0069] 111In or 99mTc, 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 present invention.
  • As modifications and changes may be made in the structure of the water soluble polymer such as a water soluble polyamino acid, or a water soluble metal chelator, of the present invention and still obtain molecules having like or otherwise desirable characteristics, such “biologically functional equivalents” or “functional equivalents” are also encompassed within the present invention. [0070]
  • For example, one of skill in the art will recognize that certain amino acids may be substituted for other amino acids in a polyamino acid structure, including water soluble amino acid polymers such as poly-glutamic acid, poly-aspartic acid, or poly-lysine, without appreciable loss of interactive binding capacity with structures such as, for example, a chemotherapeutic and/or antiangiogenic drug, such as paclitaxel or docetaxel, or such like. Additionally, amino acid substitutions in a water soluble polyamino acid conjugated to a chemotherapeutic and/or antiangiogenic drug, such as paclitaxel or docetaxel, or such like, as exemplified by but not limited to PG-TXL, may be made and still maintain part or all of the novel pharmacological properties disclosed herein. Since it is the interactive capacity and nature of a protein that defines that protein's biological functional activity, certain amino acid sequence substitutions can be made in a polyamino acid sequence and nevertheless obtain a polyamino acid with like (agonistic) properties. It is thus contemplated by the inventors that various changes may be made in the sequence of the water soluble polyamino acids of a drug conjugate, such as, but not limited to PG-TXL, without appreciable loss of their biological utility or activity. [0071]
  • In terms of functional equivalents, it is well understood by the skilled artisan that, inherent in the definition of a “biologically functional equivalent of a water soluble polyamino acid”, is the concept that there is a limit to the number of changes that may be made within a portion of the molecule and still result in a molecule with an acceptable level of equivalent biological activity. Biologically functional equivalent of a water soluble polyamino acids, are thus defined herein as those water soluble polyamino acids in which certain, not most or all, of the amino acids may be substituted by non-water soluble amino acids, whether natural, unusual, or chemically modified. [0072]
  • In particular, where shorter length water soluble polyamino acids are concerned, it is contemplated that fewer amino acids should be made within the given peptide. Longer domains may have an intermediate number of changes. The longest water soluble polyamino acid chains, as described herein, will have the most tolerance for a larger number of changes. Of course, a plurality of distinct water soluble polyamino acids, such as but not limited to poly glutamic acid, poly aspartic acid, or poly-lysine, with different substitutions may easily be made and used in accordance with the invention. [0073]
  • It is also well understood that where certain residues are shown to be particularly important to the biological or structural properties of a polyamino acid, such residues may not generally be exchanged. In this manner, functional equivalents are defined herein as those water soluble polyamino acids which maintain a substantial amount of their native biological activity. [0074]
  • Amino acid substitutions are generally based on the relative similarity of the amino acid side-chain substituents, for example, their hydrophobicity, hydrophilicity, charge, size, and the like. An analysis of the size, shape and type of the amino acid side-chain substituents reveals that arginine, lysine and histidine are all positively charged residues; that alanine, glycine and serine are all a similar size; and that phenylalanine, tryptophan and tyrosine all have a generally similar shape. Therefore, based upon these considerations, arginine, lysine and histidine; alanine, glycine and serine; and phenylalanine, tryptophan and tyrosine; are defined herein as biologically functional equivalents. [0075]
  • To effect more quantitative changes, the hydropathic index of amino acids may be considered. Each amino acid has been assigned a hydropathic index on the basis of their hydrophobicity and charge characteristics, these are: isoleucine (+4.5); valine (+4.2); leucine (+3.8); phenylalanine (+2.8); cysteine/cystine (+2.5); methionine (+1.9); alanine (+1.8); glycine (−0.4); threonine (−0.7); serine (−0.8); tryptophan (−0.9); tyrosine (−1.3); proline (−1.6); histidine (−3.2); glutamate (−3.5); glutamine (−3.5); aspartate (−3.5); asparagine (−3.5); lysine (−3.9); and arginine (−4.5). [0076]
  • The importance of the hydropathic amino acid index in conferring interactive biological function on a protein, and correspondingly a polyamino acid, is generally understood in the art (Kyte & Doolittle, 1982, incorporated herein by reference). It is known that certain amino acids may be substituted for other amino acids having a similar hydropathic index or score and still retain a similar biological activity. In making changes based upon the hydropathic index, the substitution of amino acids whose hydropathic indices are within ±2 is preferred, those which are within ±1 are particularly preferred, and those within ±0.5 are even more particularly preferred. [0077]
  • It is also understood in the art that the substitution of like amino acids can be made effectively on the basis of hydrophilicity. As detailed in U.S. Pat. No. 4,554,101, the following hydrophilicity values have been assigned to amino acid residues: arginine (+3.0); lysine (+3.0); aspartate (+3.0±1); glutamate (+3.0±1); serine (+0.3); asparagine (+0.2); glutamine (+0.2); glycine (0); threonine (−0.4); proline (-0.5±1); alanine (−0.5); histidine (−0.5); cysteine (−1.0); methionine (−1.3); valine (−1.5); leucine (−1.8); isoleucine (−1.8); tyrosine (−2.3); phenylalanine (−2.5); tryptophan (−3.4). In making changes based upon similar hydrophilicity values, the substitution of amino acids whose hydrophilicity values are within ±2 is preferred, those which are within ±1 are particularly preferred, and those within ±0.5 are even more particularly preferred. Hence, in reference to hydrophilicity, arginine, lysine, aspartic acid, and glutamic acid are defined herein as biologically functional equivalents, particularly in water soluble amino acid polymers. [0078]
  • In addition to the water soluble polyamino acid-chemotherapeutic and/or antiangiogenic drug compounds described herein, such as paclitaxel or docetaxel conjugated to a water soluble amino acid, or such like, as exemplified by, but not limited to PG-TXL compounds described herein, the inventors also contemplate that other sterically similar compounds may be formulated to mimic the key portions of the water soluble polyamino acid structure. Such compounds, which may be termed peptidomimetics, may be used in the same manner as the peptides of the invention and hence are also functional equivalents. [0079]
  • Certain mimetics that mimic elements of protein secondary structure are described in Johnson et al. (1993). The underlying rationale behind the use of peptide mimetics is that the peptide backbone of proteins, including polyamino acids, exists chiefly to orientate amino acid side chains in such a way as to facilitate molecular interactions, such as those of antibody and antigen. A peptide mimetic is thus designed to permit molecular interactions similar to the natural molecule. [0080]
  • Some successful applications of the peptide mimetic concept have focused on mimetics of β-turns within proteins, which are known to be highly antigenic. Likely β-turn structure within a polypeptide can be predicted by computer-based algorithms, as discussed herein. Once the component amino acids of the turn are determined, mimetics can be constructed to achieve a similar spatial orientation of the essential elements of the amino acid side chains. [0081]
  • The generation of further structural equivalents or mimetics may be achieved by the techniques of modeling and chemical design known to those of skill in the art. The art of receptor modeling is now well known, and by such methods a chemical that binds to water soluble polyamino acids can be designed and then synthesized. It will be understood that all such sterically designed constructs fall within the scope of the present invention. [0082]
  • In addition to the 20 “standard” amino acids provided through the genetic code, modified or unusual amino acids are also contemplated for use in the present invention. A table of exemplary, but not limiting, modified or unusual amino acids is provided herein below. [0083]
    TABLE 1
    Modified and Unusual Amino Acids
    Abbr. Amino Acids
    Aad 2-Aminoadipic acid
    bAad 3-Aminoadipic acid
    bAla beta-alanine, beta-Amino-propionic acid
    Abu 2-Aminobutyric acid
    4Abu 4-Aminobutyric acid, piperidinic acid
    Acp 6-Aminocaproic acid
    Ahe 2-Aminoheptanoic acid
    Aib 2-Aminoisobutyric acid
    bAib 3-Aminoisobutyric acid
    Apm 2-Aminopimelic acid
    Dbu
    2,4-Diaminobutyric acid
    Des Desmosine
    Dpm
    2,2′-Diaminopimelic acid
    Dpr
    2,3-Diaminopropionic acid
    EtGly N-Ethylglycine
    EtAsn N-Ethylasparagine
    Hyl Hydroxylysine
    aHyl allo-Hydroxylysine
    3Hyp 3-Hydroxyproline
    4Hyp 4-Hydroxyproline
    Ide Isodesmosine
    aIle allo-Isoleucine
    MeGly N-Methylglycine, sarcosine
    MeIle N-Methylisoleucine
    MeLys 6-N-Methyllysine
    MeVal N-Methylvaline
    Nva Norvaline
    Nle Norleucine
    Orn Orinthine
  • Toxicity studies, pharmacokinetics and tissue distribution of DTPA-paclitaxel have shown that in mice the LD[0084] 50 (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 constraints 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 maximum tolerated doses (MTD) 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. [0085]
  • 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 microtubule 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 H[0086] 2O2 production. In addition, paclitaxel may have antiangiogenic activity by blocking coordinated endothelial cell migration (Oliver et al. 1994). Therefore, the water soluble polyamino acids conjugated paclitaxel of the present invention are contemplated to be useful in the treatment of rheumatoid arthritis. The polymer conjugated formulation disclosed herein would also offer the advantages of controlled 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. [0087]
  • 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. [0088]
  • 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. [0089]
  • 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. [0090]
  • 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. [0091]
  • 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. [0092]
  • EXAMPLE 1 Poly-glutamic Acid-Paclitaxel (PG-TXL)
  • The present example concerns a first study involving the conjugation of paclitaxel to a water-soluble polymer, poly (l-glutamic acid) (PG) and the efficacy of the preparation against a variety of tumors in mice and rats. The potential of water-soluble polymers used as drug carriers is well established (Kopecek, 1990; Maeda and Matsumura, 1989). [0093]
  • Synthesis of Poly-Gutamic Acid-Paclitaxel (PG-TXL) [0094]
  • 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), Ara-C (Kato et al., 1984) and melphalan (Morimoto et al., 1984) have been conjugated to PG. However, poly-aspartic acid may be conjugated to anti-tumor drugs using the reaction scheme described herein for PG-TXL. [0095]
  • The reaction scheme is presented in FIG. 1B. Poly(l-glutamic acid) (PG) sodium salt was obtained from Sigma (St. Louis, Mo.). The polymer by viscosity had a molecular weight of 36,200, and number-average molecular weight (M[0096] n) of 24,000 as determined by low-angle laser light scattering (LALLS). Lot-specific polydispersity (Mw/Mn) was 1.15 where Mw is weight-average molecular weight. PG sodium salt (MW 34 K, Sigma, 0.35 g) was first convened to PG in its proton form. The pH of the aqueous PG sodium salt solution was adjusted to 2.0 using 0.2 M HCl. 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 N,N-dimethylformamide (DMF) (1.5 mL) was added 22 mg paclitaxel (0.026 mmol, molar ratio PG/paclitaxel=17), 15 mg dicyclohexylcarbodiimide (DCC) (0.073 mmol) and trace amount of dimethylaminopyridine (DMAP). Paclitaxel was supplied by Hande Tech (Houston, Tex.), and the purity was 99% and higher as confirmed by HPLC assay. [0097]
  • The reaction was allowed to proceed at room temperature for 12-18 h. Thin layer chromatography (TLC, silica) showed complete conversion of paclitaxel (Rf=0.55) to polymer conjugate (Rf=0, CHCl[0098] 3/MeOH=10:1). To stop the reaction, the mixture was poured into chloroform. The resulting precipitate was collected and dried in vacuum to yield 70 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-TXL conjugate was obtained by dissolving the product in 1.0 M NaHCO[0099] 3. The aqueous solution of PG-TXL was dialyzed against distilled water (MWCO 10,000) to remove low molecular weight contaminants and excess NaHCO3 salt. Lyophilization of the dialysate yielded 98 mg of product as a white powder. The paclitaxel content in this polymeric conjugate as determined by UV was 20-22% (w/w). Yield: 98% (conversion to polymer bound paclitaxel, UV). Solubility in water>20 mg paclitaxel/ml. A similar method can be used to synthesize PG-TXL with higher paclitaxel content (up to 35%) by simply increasing the ratio of paclitaxel to PG used.
  • Characterization of Poly-Glutamic Acid-Paclitaxel (PG-TXL) [0100]
  • Ultraviolet spectra were obtained on a Beckman DU-70 spectrophotometer, using the same concentration of PG aqueous solution as reference. PG-TXL showed characteristic paclitaxel absorption with λ[0101] max shifts from 228 to 230 nm. The concentration of paclitaxel in PG-TXL conjugate 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.
  • [0102] 1H-NMR spectra were recorded with GE model GN 500 (500 MHz) spectrometer in D2O. Both the PG moieties and the paclitaxel moieties were discernible. The couplings of polymer conjugated paclitaxel are too poorly resolved to be measured with sufficient accuracy. Resonances at 7.75 to 7.36 ppm are attributable to aromatic components of paclitaxel resonances at 6.38 ppm (C120-H), 5.97 ppm (C13-H), 5.63 ppm (C2′-H, d), 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 tentatively assigned to aliphatic components of paclitaxel. Other resonances 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. Although a peak at 5.63 ppm could be tentatively assigned to the C-2′ proton of the C-2′ ester, the C-2′ proton of unsubstituted paclitaxel at 4.78 ppm was also present, suggesting that the resulting conjugate may contain paclitaxel substitutions at both the C-2′ and C-7 positions. A 100 mg/ml solution of the conjugate produces a clear, viscous, yet flowable liquid. This procedure consistently produces PG-TXL conjugate containing 20% of paclitaxel by weight, i.e., approximately 7 paclitaxel molecules are bound to each polymer chain.
  • Gel Permeation Chromatography Studies of Poly-Glutamic Acid-Paclitaxel (PG-TXL) [0103]
  • The relative molecular weight of PG-TXL 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 ultraviolet (UV) detection set at 270 nm. For PG-TXL sodium salt, a TSK-gel column suitable for analysis of water-soluble polymer was used, and the system was eluted with 0.2 mM PBS (pH 6.8) at 1.0 ml/min. Conjugation of paclitaxel to PG resulted in an increase in the molecular weight of PG-TXL, as indicated by the shift of retention time from 6.4 min for PG to 5.0 min for PG-TXL conjugate. The crude product contained small molecular weight contaminants (retention time 8.0 to 10.0 min, and 11.3 min), which can be effectively removed by convening PG-TXL to its sodium salt, followed by dialysis. [0104]
  • Hydrolytic Degradation of a Poly-Glutamic Acid-Paclitaxel (PG-TXL) Conjugate [0105]
  • To gain insight on the release kinetics of paclitaxel and related molecular species from PG-TXL, the hydrolytic stability of PG-TXL was tested in PBS at various pH. High performance liquid chromatography (HPLC) revealed that incubation of PG-TXL in PBS solutions produced paclitaxel and several other species including one that is more hydrophobic than paclitaxel (metabolite 1). The fact that these species all were derived from paclitaxel was confirmed through similar degradation studies using PG-[[0106] 3H]TXL. Based on its retention time on HPLC, metabolite-l is probably 7-epipaclitaxel, a biologically active isomer of paclitaxel. In fact, the amount of metabolite 1 recovered in PBS surpassed that of paclitaxel after 5 days and 1 day of incubation at pH 7.4 and pH 9.5 respectively (FIG. 6). At pH 5.5 and pH 7.4, the release profiles of metabolite 1 indicated pseudo-zero order kinetics and displayed a delay time varying from 3 days (pH 5.5) to 7 h (pH 7.4), suggesting that metabolite-1 is a secondary product. Apparently, PG-TXL is more stable in acidic solution than in basic solution.
  • In Vivo Antitumor Activity [0107]
  • 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. [0108]
  • The tumor growth delay induced by PG-TXL was measured in mammary ovarian carcinoma (OCA-I) implanted in C3Hf/Kam mice. All tumors were syngeneic to this strain. Solitary tumors were produced in the muscle of the right thigh of female C3H/Kam mice (25-30 g) by injecting 5×10[0109] 5 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 1.0×105 viable 13762F tumor cells in 0.1 ml PBS. Treatments were initiated when the tumors in mice had grown to 500 mm3 (10 mm in diameter), or when the tumors in rats had grown to 2400 mm3 (mean diameter 17 mm).
  • PG-TXL was disolved in saline (10 mg equivalent paclitaxel/ml), and paclitaxel was dissolved in Cremophor EL(g vehicle (6 mg/ml). Data are presented as mean ± standard deviation of tumor volumes. In control studies, saline (0.6 ml), Cremophor vehicle [50/50 Cremophor/ethanol diluted with saline (1:4)], PG solution in saline, and paclitaxel plus PG were used. The maximum tolerated dose (MTD) of PG-TXL and paclitaxel in normal female C3Hf/Kam mice was estimated to be 160 mg/kg and 80 mg/kg respectively. A single dose of PG-TXL in saline or paclitaxel in Cremophor EL vehicle was given in doses varying from 40 to 160 mg equiv. Paclitaxel/kg body weight. Tumor growth was determined daily (FIGS. 7A, 7B, [0110] 7C, 7D and 7E) by measuring three orthogonal tumor diameters. Tumor volume was calculated according to formula (A×B×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 mm3 in mice minus the time in days for tumors treated with saline control to grow from 500 to 2,000 mm3. When the tumor size reached 2000 mm3, the tumor growth delay was calculated; the mice were sacrificed when tumors were approximately 2500 mm3. The PG-TXL group were (n=6 and 7), other each group were (n=5). Table 2 summarizes acute toxicity of PG paclitaxel in rats in comparison with paclitaxel/Cremophor. Table 3 summarizes the data concerning the effect of PG-TXL against MCa-4, FSa-II and HCa-I tumors in mice. The data are also summarized in FIG. 7A-FIG. 7E.
    TABLE 2
    Acute Toxocity of PG-TXL in Fischer Rats*
    # of Body Time of Full
    Dose Toxic Weight Time at Nadir Recovery
    Group (mg/kg) Death Loss in % (days) (days)
    PG-TXL a 60 1/4 15.7 7 14
    PG-TXL a 40 0/4 11.1 6 11
    Paclitaxel b 60 1/4 16.7 6 15
    Paclitaxel b 40 0/3 17.9 6 16
    Paclitaxel b 20 0/5 17.0 5 N/A
  • [0111]
    TABLE 3
    The Antitumor Effect of PG-TXL Against
    Different Types of In vivo Murine Tumors
    Time
    to Growbb
    Tumor Druga 500-2000 mm3 AGDc t-testd
    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-TXL (40 mg/kg) 8.6 ± 1.2 (4) 3.8 0.026
    PG-TXL (60 mg/kg) 14.2 ± 1.1 (5)  9.4 0.0001
    PG-TXL (120 mg/kg) 44.4 ± 2.9 (5)  39.6 <0.0001
    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-TXL (80 mg/kg) 3.8 ± 0.4 (6) 1.9 0.0016
    PG-TXL (160 mg/kg)  5.1 ± 0.3 (13) 3.2 <0.0001
    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-TXL (40 mg/kg) 8.2 ± 0.7 (5) 0.9 0.218
    PG-TXL (80 mg/kg) 8.6 ± 0.2 (5) 1.3 0.0053
    PG-TXL (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
  • Two important findings emerged from these studies. First, like paclitaxel, there is an intertumor variability of the antitumor effect of water-soluble PG-TXL. PG-TXL is most effective against MCa-4 and OCA-1 tumors. Second, PG-TXL is more effective than paclitaxel on equivalent mg paclitaxel basis in the case of MCa-4, HCa-I, and on OCA-1 tumors, and is remarkably potent at its maximum tolerated dose (MTD). [0112]
  • In a parallel study, the antitumor activity of PG-TXL in Fischer rats with the well established rat [0113] mammary adenocarcinoma 13762F was examined. Female Fischer 344 rats (125-150 g) were injected with 1.0×105 viable 13762F tumor cells in 0.1 ml PBS. Once tumors reached a mean volume of 2000 mm3 (mean diameter, 1.6 cm), animals were treated using a similar protocol as described above. Tumor growth was determined daily by measuring three orthogonal tumor diameters. Tumor volume was calculated according to the formula (A×B×C)/2. A single dose of PG-TXL in saline or paclitaxel in a Cremophor EL® vehicle was given in doses varying from 20 to 60 mg equivalent paclitaxel/kg body weight. In control studies, saline, the Cremophor EL® vehicle [50/50 Cremophor/ethanol diluted with saline (1:4)], PG solution in saline and paclitaxel plus PG were used. Again, complete tumor eradication at the MTD of PG-TXL (60 mg equivalent paclitaxel/kg) was observed. PG-TXL given at a lower dose of 40 mg equivalent paclitaxel/kg also resulted in complete tumor regression (FIG. 7B). In contrast, the MTD of paclitaxel in Cremophor EL® was less than 20 mg/kg. Paclitaxel at this dose caused a tumor growth delay (Tumor growth delay is defined as the time in days for tumors treated with the test drugs to grow from 2,000 mm3 to 10,000 mm3 minus the time in days for tumors treated with saline control to grow from 2,000 mm3 to 10,000 mm3.) of only 5 days, whereas the same equivalent paclitaxel dose of PG-TXL resulted in a tumor growth delay of 23 days (FIG. 7B).
  • Studies of Nude Mice Injected with Human Breast Cancer and Treated with PG-TXL [0114]
  • Nude mice were injected with 2×10[0115] 6 MDA-435-Lung2 cells (a variant of the MDA-MB-435 human breast cancer cell line) into the mammary fatpad. When the tumors reached 5 mm mean diameter, (27 days after tumor injection), mice were treated with an i.v. injection of PG-TXL or the various controls (see Table 4). Tumor measurements were taken weekly. Tumors that reached 1.5 cm were removed surgically. All mice were killed at 120 days, and remaining tumors removed and weighed. Mice were examined for metastases, and lungs processed for histology, with single sections of the organs scored for the presence of micrometastases.
    TABLE 4
    No.
    Tumor Mean tumor tumors Lung
    Treatment takea wt (g)b regressedc metastasesd
    PBS 5/6  1.3 ± 0.24 4/5 (80%)
    Cremophor 9/9 1.26 ± 0.67 4/8 (50%)
    PGA 10/10 1.13 ± 0.7  4/7 (57%)
    Taxol ™/Cremophor 10/10 1.31 ± 0.69 3/7 (42%)
    60 mg/kg
    PG-TXL 60 mg/kg 10/10 1.23 ± 0.38 2/10   5/8 (62.5%)
    PG-TXL 120 mg/kg  9/10 0.925 ± 0.12  4/8  1/4 (25%)
    #equivalent of PG-TXL.
  • From the results of the study in which a single bolus of PG-conjugated paclitaxel (PG-TXL) was given, at a drug equivalent of 120 mg/kg paclitaxel, it is apparent that the MDA-435 cancer cell line responds to the drug and that this formulation of the drug is much better tolerated than when Cremophor is the vehicle. [0116]
  • In the breast cancer study using MDA-MB-435, only the higher dose of PG-TXL inhibited the growth rate of the mammary fatpad tumors. From the growth curve it was apparent that tumor growth resumed approximately 30 days after the single dose of conjugate. However, the growth curve does not reveal that in the PG-TXL 120 mg/kg group there were a number of tumor regressions. As shown in Table 3, the incidence of lung metastasis in the mice with residual tumors was also reduced. While the numbers of mice in the study are small, they do suggest that the therapy was effective in reducing both local tumor growth and incidence of metastasis. [0117]
  • In this study design it is not possible to distinguish whether a lower incidence of metastasis is due to a reduction of tumor mass of the primary site, or due to a direct effect on any micrometastases that may have already been established at the time of therapy. [0118]
  • In Vivo Therapy of Human Breast Cancer Using Multiple Injections of PG-TXL [0119]
  • To test the effect of multiple injections of PG-TXL, nude mice were injected with 2×10[0120] 6; MDA-435-Lung 2 cells (a variant of the MDA-MB-435 human breast cancer cell line) into the mammary fatpad. When the tumors reached 5 mm mean diameter, the treatments were started, and repeated at 14 day intervals (day 24, 38, 52) for a total of three injections. Tumor measurements were taken weekly. The mice were killed on day 105 after tumor cell injection, and the tumor weights and incidence of metastasis recorded. The lungs were processed for histology, and single sections scored for the presence of micrometastases. The results are shown in Table 5.
    TABLE 5
    Tumors
    Treatment Tumor takea Mean weight (g)b regressedc Metastasisd
    None 4/5 1.83 ± 0.15 4/4 (100%)
    PG-control  6/10  1.7 ± 0.11 5/6 (83%) 
    PG-TXL/60  7/10 1.36 ± 0.28 6/7 (86%) 
    mg
    PG-TXL/120  8/10 0.97 ± 0.22 p = 2/8 2/6 (33%) 
    mg 0.011e
  • In Vivo Therapy of Human Ovarian Cancer Using PG-TXL Conjugate [0121]
  • Nude mice were injected i.p. with the human ovarian cancer cell line, SKOV3ip1. Five days after tumor injection, the mice were injected i.v. with the PG-paclitaxel (PG-TXL), at concentrations equivalent to 120 mg/kg or 160 mg/kg of paclitaxel. Initially the plan was to repeat these injections at 7-day intervals, but a single injection of the 160 mg/kg dose killed 5 of the 10 mice. Only the 120 mg/kg group received three injections. The study was terminated on day 98, and any surviving mice killed. The results are shown in FIG. 14, and in Table 6. [0122]
  • The median survival values for the groups at present are: untreated=47 days, PC-control=43 days, PG-TXL (120 mg/kg)=83 days, PG-TXL (160 mg/kg)=83 days [note that this does not include the mice that died from the initial toxicity of the drug]. [0123]
    TABLE 6
    Median
    Treatment Tumor takea survival (range)b Ascitcsc Mean vol (ml)d
    None 10/10 56 (38-98)  8/10 2.2 ± 1.6
    PG-control 8/9 45 (39-98)  8/8  2.2 ± 1.6
    PG-120 7/8 82 (59-98)  3/7  2.7 ± 1.4
    PG-160 3/5c 84 (34f-98) 0/3 
  • The PG-TXL 120 mg/kg significantly extended the survival of the mice with intraperitoneal SKOV3ipl, (a human ovarian cancer cell line which overexpresses HER2/neu), compared with mice injected with PG alone. Multiple doses and/or increasing the dose of conjugate may significantly reduce the tumor incidence in addition to extending survival. [0124]
  • In the nude mice studies above, the growth curves show that although breast cancer growth is checked by paclitaxel, especially with the higher dose conjugated with PG, tumor size continues to increase about a month after the therapy. A second (or third) round of therapy may have caused the tumor growth to plateau, or give more tumor regressions. The growth curves do not include the tumors that regressed—as shown in Table 4, the tumors shrank/disappeared in 50% of the mice treated with the highest dose of PG-TXL, and of the 4 animals with progressively growing tumors at the end of the study, only one had micrometastases in the lungs. So the treatment that reduced growth of the primary tumors also reduced the incidence of metastasis. The incidence of metastasis in all other therapy groups, including the control groups of Cremophor and PG were lower than the PBS control, therefore it is probably not valid to state that the reduction in incidence of metastasis in the Taxol™/Cremophor group is a significant finding. [0125]
  • EXAMPLE 2 DTPA-Paclitaxel
  • Synthesis of DTPA-Paclitaxel: [0126]
  • To a solution of paclitaxel (100 mg, 0.1 17 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 C[0127] 18 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 1H NMR spectrum (DMSO-d6) the resonance of NCH2CH2N and CH2COOH 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 NaHCO[0128] 3, followed by lyophilizing to yield a water-soluble solid powder (solubility>20 mg equivalent paclitaxel/ml).
  • Hydrolytic Stability of DTPA-Paclitaxel: [0129]
  • 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[0130] 3 aqueous solution (pH 9.3) and analyzed by HPLC. The HPLC system consisted of a Waters 150×3.9 (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 B16 Mouse Melanoma Cells In Vitro: [0131]
  • Cells were seeded in 24-well plates at a concentration of 2.5×10[0132] 4 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 h 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 5×10−9 M to 75×10−9 M. After 40 h, 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 IC[0133] 50 for paclitaxel and DTPA-paclitaxel are 15 nM and 7.5 nM, respectively.
  • Antitumor Effect on Mammary Carcinoma (MCa-4) Tumor Model: [0134]
  • Female C3Hf/Kam mice were inoculated with mammary carcinoma (MCa-4) in the muscles of the right thigh (5×10[0135] 5 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). [0136]
  • Radiolabeling of DTPA-Paclitaxel with [0137] 111In
  • 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 [0138] 111InCl3 solution (1.0 mCi) in sodium acetate buffer (pH 5.5). After an incubation period of 30 min at room temperature, the labeled 111In-DTPA-paclitaxel was purified by passing the mixture through a C18 Sep-Pac cartridge using saline and subsequently ethanol as the mobile phase. Free 111n-DTPA (<3%) was removed by saline, while 111In-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 [0139] 111In-DTPA-Paclitaxel:
  • HPLC was used to analyze the reaction mixture and purity of [0140] 111In-DTPA-paclitaxel. The system consisted of a LDC binary pump, a 100×8.0 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 NaI crystal detector and a Spectra-Physics UV/Vis detector. As evidenced by HPLC analysis, purification by Sep-Pak cartridge removed most of the 111In-DTPA, which had a retention time of 2.7 min. The 111In-DTPA was probably derived from traces of DTPA contaminant in the DTPA-paclitaxel. A radio-chromatogram of 111In-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: [0141]
  • Female C3Hf/Kam mice were inoculated with mammary carcinoma (MCa-4) in the muscles of the right thigh (5×10[0142] 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 111In-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 h after injection. In group II, the same procedures were followed except that the mice were injected with 111In-DTPA as a control. FIG. 5 shows gamma-scintigraphs of animals injected with 111In-DTPA and 111In-DTPA-paclitaxel. 111In-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, 111In-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 111In-DTPA-paclitaxel was also excreted through kidney, which only played a minor role in the clearance of paclitaxel. The tumor had significant uptake of 111In-DTPA-paclitaxel. These results demonstrate that 111In-DTPA-paclitaxel is able to detect certain tumors and to quantify the uptake of 111In-DTPA-paclitaxel in the tumors, which in turn, may assist in the selection of patients for the paclitaxel treatment. In contrast, the smaller PG-TXL conjugate has a different distrubution than DTPA-paclitaxel, and partly localizes in the liver and tumors of test animals.
  • EXAMPLE 3 Polyethylene glycol-Paclitaxel
  • Synthesis of Polyethylene Glycol-Paclitaxel (PEG-Paclitaxel) [0143]
  • 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 h. 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 [0144] mg 2′-succinyl-paclitaxel. PEG-paclitaxel was synthesized by an N-ethoxycarbonyl-2-ethoxy-1,2-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 h. The crude product was chromatographed on silica gel with ethyl acetate followed by chloroform-methanol (10:1). This gave 350 mg of product. 1H NMR (CDCl3) 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 [0145]
  • 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. [0146]
  • Cytotoxicity Studies of PEG-Paclitaxel Using B16 Mouse Melanoma Cells In Vitro [0147]
  • Following the procedure described in the cytotoxicity studies with DTPA-paclitaxel, melanoma cells were seeded in 24-well plates at a concentration of 2.5×10[0148] 4 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 h in a 97% humidified atmosphere of 5.5% CO,. The medium was then replaced with fresh medium containing paclitaxel or its derivatives in concentrations ranging from 5×10−9 M to 75×10−9 M. After 40 h, 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 5×10−9 M to 75×10−9 M also did not effect cell proliferation.
  • Antitumor Effect of PEG-Paclitaxel Against MCa-4 Tumor in Mice [0149]
  • To evaluate the antitumor efficacy of PEG-paclitaxel against solid breast tumors, MCa-4 cells (5×10[0150] 5 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 min of injection. PEG-paclitaxel was dissolved in saline (6 mg equiv. paclitaxel/ml) and filtered through a sterile filter (Millipore, 4.5 elm). 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 studies. 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 (ρ>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). [0151]
  • EXAMPLE 5 Poly(L-glutamic acid)-Paclitaxel (PG-TXL) and Paclitaxel Pharmacological Properties
  • The objective of this study was to compare PG-TXL and paclitaxel pharmacological properties in their ability to promote in vitro assembly of tubulin, to inhibit cell growth against rat mammary [0152] tumor cell line 13762F and human breast tumor cell lines, to induce p53 protein, and to rescue a paclitaxel-dependent mutant cell line. Paclitaxel's release from PG-TXL in vivo was measured to determine if PG-TXL's mechanism of action can be attributed to free pacitaxel.
  • Microtubule Polymerization Using Poly-Glutamic Acid-Paclitaxel (PG-TXL) and Paclitaxel [0153]
  • To test whether intact PG-TXL has any intrinsic biological activity in promoting tubulin polymerization, paclitaxel, PG-TXL, and “aged” PG-TXL were compared for their relative ability to promote in vitro assembly of purified bovine brain tubulin. The tubulin assembly reaction was performed at 32° C. in PEM buffer (80 mM PIPES buffer, 1 mM EGTA, 1 mM MgCl[0154] 2, pH 6.9) at a tubulin (bovine brain, Cytoskeleton Inc., Boulder, Colo.) concentration of 1 mg/ml (10 μM) in the presence or absence of drugs (1.0 μM equivalent paclitaxel) and 1.0 mM guanosine 5′-triphosphate (GTP). “Aged” PG-TXL was obtained by placing PG-TXL in PBS (pH 7.4) at 37° C. for 3 days. Tubulin polymerization was followed by measuring the absorbance of the solution at 340 nm over time. Addition of 1 μM paclitaxel to a solution of tubulin in assembly buffer caused a clear increase in absorbance due to the increase in light scattering resulting from the polymerization of tubulin into microtubules. In contrast, a 10 λM paclitaxel equivalent of PG-TXL had no effect on polymerization. A solution of the conjugate that was “aged” for 3 days in PBS (pH 7.4) at 37° C. exhibited enhanced activity although its ability to promote tubulin polymerization was still markedly less than paclitaxel (FIG. 10).
  • Effects of Poly-Glutamic Acid-Paclitaxel (PG-TXL) on the Growth of Rat and Human Tumor Cell Lines In Vitro [0155]
  • To evaluate whether the superior antitumor activity of PG-TXL observed in animals is due to increased cytotoxicity, PG-TXL and paclitaxel were compared for their ability to inhibit cell growth against the established rat mammary [0156] tumor cell line 13762F. The effect of PG-TXL on cell growth was examined by a plating efficiency assay. Rat 13762F cells were seeded (200 cells) into 60 mm dishes containing drug concentrations varying from 0 to 200 nM in growth medium (cc modified minimum essential medium [α-MEM] containing 5% fetal bovine serum, 50 U/ml of penicillin, and 50 μg/ml of streptomycin). After 6 days of growth, the cells were stained with a 0.1% methylene blue solution and colonies were counted. The drug concentration producing 50% inhibition of colony formation (IC50) was then calculated. The approximate IC50 values after 6 days of continuous exposure were: paclitaxel (2 nM), PG-TXL (100 nM), “aged” (see below) PG-TXL (50 nM). It is clear that PG-TXL is approximately 30-50 fold less potent than paclitaxel itself. When PG-TXL was incubated in phosphate buffered saline solution (PBS, pH 74) at 37° C. for 3 days to obtain an “aged” solution, only about 10% of paclitaxel was released. Since the “aged” solution is more potent than freshly dissolved PG-TXL, the in vitro degradation of PG-TXL or release of active drug appears to be important for PG-TXL to exert this biological activity. However, even after “aging,” PG-TXL is still 25 times less potent than paclitaxel.
  • In a similar study, the effect of PG-TXL on cell growth of human breast cancer cell lines was examined by MTT assay after 3 days of continuous exposure. While PG-TXL was 8-and 30-fold more potent than paclitaxel against MDA330 and MDA-MB453 cell lines, PG-TXL was 2- and 3-fold less potent than paclitaxel against MCF7/her-2 and MCF7 cell lines. These results suggest that PG-TXL and paclitaxel have different activity against different cell lines. PG-TXL may be a product with distinct pharmacological properties different from that of paclitaxel. [0157]
  • The Ability of Poly-Glutamic Acid-Paclitaxel (PG-TXL) to Support a Paclitaxel-Dependent Cell Line In Vitro [0158]
  • The inventors investigated the ability of PG-TXL to rescue a paclitaxel-dependent mutant cell line. [0159] Tax 18, a CHO cell line selected for resistance to paclitaxel, is a well characterized mutant that has been found to require the continuous presence of paclitaxel for cell division. In the absence of drug, a functional mitotic spindle apparatus is unable to form (Cabral et al., 1983). The mitosis phase of the cell cycle is prolonged with subsequent failure to segregate chromosomes and to divide into daughter cells. Nonetheless, the cells continue to progress through the cell cycle and replicate their DNA resulting in the formation of large polyploid cells that eventually die due to genomic instability (Cabras and Barlow, 1991). Low concentrations of paclitaxel are able to rescue the mutant phenotype by permitting microtubule assembly and the formation of sufficient mitotic spindle fibers. Thus, these cells provide a convenient bioassay for agents that promote microtubule assembly. Growth of paclitaxel-dependent CHO mutant Tax-18 cells was carried out on 24-well tissue culture dishes. Approximately 100 cells were added to wells containing growth medium and equivalent concentrations of paclitaxel varying from 0 to 1.0 μM. After 6 days of incubation at 37° C., the medium was removed and the cells were stained with methylene blue.
  • Little or no increase in cell number is seen in the absence of drug, but concentrations of paclitaxel between 0.05-0.2 μM clearly support the growth of this cell line. Higher concentrations of paclitaxel are presumably toxic to the cells because of overstabilization of the microtubules as is observed for normal cells. On the other hand, freshly prepared PG-TXL shows little ability to rescue Tax-18 cell growth even at the highest paclitaxel-equivalent concentration tested (1 μM). When PG-TXL was “aged” by incubating in PBS for 6 days at 37° C., its ability to support Tax-18 cell growth was partially restored. These data indicate that PG-TXL does not promote microtubule assembly, and that the in vitro biological activity of “aged” PG-TXL is a contribution of paclitaxel released from poly-glutamic acid-paclitaxel (PG-TXL). [0160]
  • The Release of [[0161] 3H]paclitaxel from PG-[3H]TXL In Vivo
  • To assess the pharmacokinetic and release characteristics of paclitaxel in vivo, normal female C3Hf/Kam mice (25-30 g) were injected with a dose of 20 mg equivalent [[0162] 3H]paclitaxel or PG-[3H]paclitaxel intravenously into the tail vein. Each mouse received 6 μCi of radiolabeled drug. [3H]paclitaxel was dissolved in Cremophor EL® vehicle whereas PG-[3H]paclitaxel was dissolved in saline. Volume injected into each mice was between 0.2 to 0.3 ml. At 0, 5, 15, 30 min, and 1, 2, 4, 8, 16, 24, 48 h postinjection, animals were sacrificed and blood samples were collected (4-5 mice per time point). Total radioactivity in plasma was measured by liquid scintillation counting (Beckman Model LS 6500, Fullerton, Calif.) using 10 μl aliquots of plasma. Up to 200 μl plasma was extracted with 3 volume of ethyl acetate according to Longnecker et al. (1987). The extraction efficiency for paclitaxel was 80%. The samples were centrifuged for 5 min at 2500 rpm, and the supernatant was separated and brought to dryness. The dried extract was reconstituted with 195 μl of HPLC mobile phase, mixed with 5 μl of cold paclitaxel (0.2 mg/ml), and 100 μl was injected onto the HPLC for determination of free paclitaxel radioactivity. Pharmacokinetic parameters were analyzed by a noncompartmental model using the WinNonlin software package. Each data point generated was the mean value of 4-5 mice.
  • The clearance of both drugs from plasma is shown in FIG. 11. While paclitaxel has an extremely short half life in plasma of mice (t[0163] ½, 29 min), the apparent half life of PG-TXL is prolonged (t½, 317 min). Slower clearance of PG-TXL from the blood was a design feature of the polymer-drug conjugate with the goal of improving tumor uptake. Surprisingly, the rate of conversion of PG-TXL to paclitaxel in plasma is slow with less than 0.1% of the radioactivity from PG-[3H]TXL being recovered as [3H]paclitaxel within 144 h after drug injection (FIG. 11).
  • In a separate study, mice bearing OCA-1 tumors were prepared as described previously. When the tumor reached 500 mm[0164] 3, animals were injected with a dose of 20 mg equivalent paclitaxel/kg of [3H]paclitaxel or PG-[3H]TXL into the tail vein. Animals were killed at 2, 5, 9, 24, 48, and 144 h postinjection. Tumors were removed, weighed, and homogenized with 3 volume of PBS (w/v). Percent of injected dose per gram tissue is calculated based on total radioactivity associated with the tumor, which was determined by counting prepared tissue homogenate aliquots. An aliquot of tissue homogenate was mixed with tissue solubilizer, followed by addition of scintillation solvent, and counted for total radioactivity. The counting efficiency was verified by the method of standard addition. Alternatively, aliquots of tissue homogenates were extracted with ethyl acetate and analyzed for free paclitaxel by HPLC. The HPLC system consisted of a 150×3.9 mm Nova-Pak column (Waters, Milford, MA), a liquid chromatography pump (Waters model 510), a UV/Vis detector set at 228 nm (Waters model 486), a flow scintillation analyzer (Packard model 500TR, Downers Grove, Ill.), and a Packard radiomatic software for data analysis. The eluting solvent (methanol:watter=2:1) was run at 1.0 ml/min. The uptake of total drugs in OCA-1 tumor was expressed as a percentage of the administered dose per gram of tissue and the association of radioactivity within OCA-1 tumor as free paclitaxel was expressed as dpm per gram tissue.
  • Quantitative assessment of tumor uptake in C3Hf/Kam mice showed that relatively high levels of radioactivity from radiolabeled PG-TXL appear in tumor tissue shortly after injection (FIG. 12A) as compared to radiolabeled paclitaxel. However, only small amounts of radioactivity within tumor tissue are due to the release of free paclitaxel (FIG. 12B). Data are presented in FIG. 12A and FIG. 12B as mean±SD from 3 mice per time point. The percent of radioactivity within tumor tissue due to paclitaxel does not appreciably increase with time suggesting that PG-TXL is not simply a prodrug for the gradual release of paclitaxel. [0165]
  • In contrast to paclitaxel, in vitro studies with PG-TXL whether prepared as a fresh solution or even after “aging” in buffer have clearly shown that the complex is not a potent cytotoxic species. It neither strongly supports tubulin polymerization nor the growth and survival of a paclitaxel-dependent CHO cell line. Furthermore, data obtained from pharmacokinetic studies indicate that both the extent and rate of release of paclitaxel in plasma is very low (less than 0.1% in 144 h). While the uptake of PG-TXL material was some 5-fold greater than that achieved by paclitaxel when using equivalent antitumor doses, that material which gains entry into tissues exists in the tissue mainly in form(s) which have been shown not to be free paclitaxel. [0166]
  • EXAMPLE 6 Effect of Polymer Structure on Activity of Water soluble polyamino Acid-Paclitaxel Conjugates
  • The present study evaluated whether antitumor activities of polymer-paclitaxel conjugates were affected by the structure of polyamino acids used for drug conjugation. Paclitaxel was coupled to poly(l-glutamic acid), poly(d-glutamic acid), and poly(l-aspartic acid) according to previously described procedures. These polyamino acid-paclitaxel conjugates had similar paclitaxel content, aqueous solubility, and molecular weight (30-40K). In C3Hf/Kam mice bearing murine OCA-1 ovarian cancer (500 mm[0167] 3 at time of treatment), a single i.v. injection of poly(l-glutamic acid)-paclitaxel at 80 mg equiv. paclitaxel/kg body weight produced a tumor growth delay of 21 days vs. saline treated controls. Poly(d-glutamic acid)-paclitaxel was as effective as poly(l-glutamic acid)-paclitaxel. However, paclitaxel conjugated with poly(l-aspartic acid) was completely inactive against OCA-1 tumor. In a separate study, the antitumor activity of polymer-paclitaxel conjugates of different molecular weight (1K, 13K, and 36K) was compared. Conjugates of lower molecular weight were significantly less effective than conjugate of higher molecular weight. The higher molecular weights above 50,000 was too viscous.
  • EXAMPLE 7 Poly-glutamic Acid-Paclitaxel (PG-TXL) Induces less Apoptosis Compared to Paclitaxel
  • To assess the mechanism of PG-TXL associated antitumor activity, histological sections of OCA-1 tumors excised from paclitaxel and PG-TXL treated mice were examined. OCA-1 tumor bearing mice were prepared as previously described. When tumor volume reached 500 mm[0168] 3, animals were injected with either paclitaxel (80 mg/kg) or PG-TXL (160 mg equivalent paclitaxel/kg). At different times ranging from 0 to 144 h after treatment, tumors were histologically analyzed to quantify mitotic and apoptotic activity according to Milas et al. (1995). The mice were killed by cervical dislocation and the tumors were immediately excised and placed in neutral-buffered formalin. The tissues were then processed and stained with hematoxylin and eosin. Both mitosis and apoptosis were scored in coded slides by microscopic examination at 400×magnification. Five fields of nonnecrotic areas were randomly selected in each histological specimen, and in each field the number of apoptotic nuclei and cells in mitosis were recorded as numbers per 100 nuclei and were expressed as a percentage. The values were based on scoring 1500 nuclei obtained from 3 mice per time point.
  • The changes observed in the paclitaxel-treated mice were qualitatively similar to those previously described (Milas et al., 1995). The tumor cells showed marked nuclear fragmentation with formation of apoptotic bodies, which was especially marked on day 1 (FIG. 13). Viable tumor cell clumps with normal mitoses were still present in these tumors by 144 h, indicating that these tumors would eventually regrow. Treatment with PG-TXL only resulted in a mild increase in mitotically arrested cells and apoptotic cells, presumably due to the small amount of free paclitaxel released from PG-TXL (FIG. 13). By 96 h, tumors from PG-TXL-treated mice developed extensive edema and necrosis, and only a small rim of viable tumor cells remained. By 144 h, the residual tumor clumps as compared to controls were comprised of cells that were larger, more pleomorphic, and that displayed less mitotic activity. [0169]
  • These data suggest that the water-soluble PG-TXL conjugate of the present disclosure has superior antitumor efficacy with reduced toxicity as compared to conventional free paclitaxel preparations. Although originally designed as a water-soluble form of paclitaxel, it is now clear that the agent used to solubilize paclitaxel, contributes to the overall anti-tumor activity of this remarkable new complex. These data indicate that PG-TXL has an ability to produce cell death in a manner which is separate from and in addition to the apoptosis produced by released free paclitaxel. [0170]
  • EXAMPLE 8 Synthesis of Poly-Glutamic Acid-Camptothecin (PG-CPT) Conjugate.
  • The synthesis of PG-CPT followed a similar reaction as previously described for the synthesis of PG-TXL. Into 80 mg of PG polymer in 2.5 ml dry DMF was added 20 mg CPT (Hande Tech.), 34 mg DCC, and trace amount of DMAP as catalyst. After stirred at room temperature overnight, the reaction mixture was poured into chloroform, and the precipitate collected. The dried precipitate was redissoved in sodium carbonate solution, dialyzed against 0.05 M phosphate buffer (pH4.5), filtered, and lyophilized. The content of CPT in the polymer conjugate was determined by fluorescence spectrometer (Perkin-Elmer Model MPF-44A) using emission wavelength of 430 nm and excitation wavelength of 370 nm. Content: 2% to 5% (w/w), solubility: >200 mg conjugate/ml. [0171]
  • EXAMPLE 9 Synthesis of Poly-Lysine (PL) TXL Conjugate (PL-TXL).
  • All accessible amine functional groups of poly-lysine (MW>30,000, Sigma) will be converted to carboxylic acid functional groups by reacting poly-lysine with succinic anhydride, glutaric anhydride, or DTPA. The remaining unreacted NH2 group in poly-lysine will be blocked by reacting the modified polymer with acetic anhydride. TXL, docetaxel, other taxiods, etopside, teniposide, camptothecin, epothilone or other anti-tumor drugs will be conjugated to the resulting polymer according to previously described procedures for the synthesis of PG-TXL. [0172]
  • EXAMPLE 10 Synthesis of Other Polyamino Acids to be Used to Conjugate TXL
  • Polyamino acid copolymers containing glutamic acid may be synthesized by the copolymerization of N-carboxyanhydrides (NCAs) of corresponding amino acid with gamma-benzyl-L-glutamate NCA. The resulting benzyl glutamate-containing copolymer will be converted to glutamic acid-containing copolymer by removing the benzyl protecting group (FIG. 15). TXL, docetaxel, other taxiods, etopside, teniposide, camptothecin, epothilone or other anti-tumor drugs will be conjugated to the resulting polymer according to previously described procedures for the synthesis of PG-TXL and PG-CPT. [0173]
  • EXAMPLE 11 Use of PG-TXL in Humans
  • Introduction [0174]
  • Poly-L-glutamic acid-Paclitaxel (PG-TXL) is a conjugate of poly-L-glutamic acid and paclitaxel. This compound is water soluble and based on early animal studies it appears that it can be administered as a short, that is several minute, intravenous injection. Based on the in vitro and early animal work, it appears that this compound is at least as active against cancer as the monomeric paclitaxel in Cremophor and may have fewer side effects. Based on these observations, this drug will be studied in humans. The study will first require formulation of this compound in a solvent which is commonly used for intravenous infusion. The inventors expect that either normal saline, 5% dextrose in water or sterile water will be used as the solvent. This formulation of PG-TXL will then be administered to at least two species of test animals such as rats and dogs to determine the toxicities of the drug in those animals and to determine a dose of the drug which then can serve as the lowest starting dose for a Phase I human study. That Phase I human study will define a dose of PG-TXL which may be used in subsequent Phase II studies in patients. Phase II studies will be performed in several tumor types to determine the activity of PG-TXL in various cancers. One of ordinary skill in the art will recognize that modifications in administration, selection of animal models and dose regiments may be made in the methods disclosed in following example, and such modifications are encompassed by the invention. [0175]
  • Animal Studies [0176]
  • These studies will be performed in rats and Beagle dogs with approximately 3 animals studied at each dose level of the drug. The levels will be increased until life threatening toxicity is noted. The animals will undergo blood testing as well as necropsy to determine the organ systems which are susceptible to this drug's toxicity and therefore to expect the side effects in human studies. Once the dose is determined which causes the death of 10% of animals then the equivalent of one-tenth of that dose will be recommended as the starting dose for human studies. This is the usual recommendation by the Food and Drug Administration (FDA) as the initial dose for human Phase I studies. [0177]
  • Phase I Studies [0178]
  • Phase I study of this drug will be performed using the starting dose defined in animal studies. The drug will be injected into the vein by a syringe over several min or alternatively it may be infused as a short infusion, up to approximately 10 to 15 min. The volume of the solvent will be from 10 ml to approximately 100 ml depending on which of the two intravenous injection approaches are used. The drug will be administered every 3 wk. This schedule is based on the early animal studies and on the schema used with paclitaxel in Cremophor. Three patients will be started on the lowest dose level as defined by the animal studies and will be treated with an injection of PG-TXL. Blood tests will be performed at baseline and weekly to evaluate blood counts; tests of liver function and renal function will be performed every 3 wk. It is expected that the counts and physiological parameters will recover sufficiently from the PG-TXL to resume the next cycle of [0179] treatment 3 wk after the previous one. If this is the case then the treatment will be repeated every 3 wk. If the first cohort of three patients tolerates the drug for 3 wk then these patients will be allowed to have the dose increased by a predetermined schema that is usually used in the Phase I studies. Once three patients have tolerated the first cycle, the next cohort of 3 patients will be started on the next higher dose level. This process of increasing the dose level will continue until at least 2 out of 3 patients at a dose level have side effects which are so severe that they prohibit continuing administration of the drug. In such a circumstance the dose level just prior to the excessively toxic one will be considered the level of drug to be administered in subsequent studies. Six to ten patients shall be treated on the dose level which will be recommended for Phase II studies to confirm its tolerability. Once the appropriate dose has been defined and acute toxic side effects of the drug evaluated, Phase II studies will be initiated.
  • Phase II Studies [0180]
  • Phase II studies of PG-TXL will be performed in several tumor types. Each study will be designed in a usual standard Phase II manner following either Gahan's or Simon's design. In brief, approximately 14 patients of a given tumor type will be treated initially, if there is no evidence of anti-cancer activity in that tumor type then further studies of PG-TXL in that tumor type will be aborted. However, if at least one patient has clinical benefit, defined as at least 50% decrease in the sum of products of perpendicular cross-sectional diameters of the tumors, then the number of patients with that tumor type treated with PG-TXL will be increased to 30. These studies will allow us to define the activity of PG-TXL in various cancers and refine the information on the side effects of the drug. The tumor types of special interest for PG-TXL will be the ones which have shown good response to paclitaxel and docetaxel. This will include ovarian cancer, breast cancer, and lung cancer. Studies comparing poly-glutamic acid-paclitaxel to paclitaxel in tumors showing response to PG-TXL will be performed. Such studies are called Phase III studies. [0181]
  • Phase III Studies of PG Paclitaxel [0182]
  • Based on the activity of paclitaxel in ovarian cancer, breast cancer, and lung cancer these will be the tumor types in which PG-TXL will be compared to paclitaxel. In view of the necessity to have a large number of patients in such randomized studies, the inventors expect that a multi-institutional study will be necessary. The inventors have in their institution access to Cooperative Community Oncology Program (CDDP) and to many other multi-institutional study groups. In addition to the potential clinical benefit of PG-TXL vs. paclitaxel, it would be appropriate to evaluate the economic impact of the two drugs. It is expected that a short term infusion of PG-TXL may result in a less costly treatment. And, therefore, there is an expectation that PG-TXL may be cost effective relative to paclitaxel monotherapy. Not only is the infusion going to be shorter, it is expected that in view of the absence of Cremophor fewer side effects will be experienced by the patients and therefore the premedication regiment including steroids and intravenous H2 and H1 blockers may no longer be necessary. All of these factors will result in a reduction in the cost of the treatment. [0183]
  • Summary [0184]
  • It is expected that the initial animal toxicology evaluation will require up to 6 months. Subsequent to that, if a drug formulation is available, human Phase I studies may be completed in another 6 to 9 months. Once these have been completed, Phase II studies in various tumor types may take another 6 to 9 months. At that point, the inventors will have a good idea of the efficacy of this drug and targeted Phase III studies may be designed and initiated. It is also possible that the Phase II studies will show enough clinical activity that abbreviated Phase III studies or no Phase III studies would be necessary. [0185]
  • 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. [0186]
  • 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. [0187]
  • Bartoni and Boitard. “In vitro and in vivo antitumoral activity of free, and encapsulated taxol,” [0188] J. Microencapsulation, 7:191-197, 1990.
  • Berstein, et al., “Higher antitumor efficacy of daunomycin when linked to dextran: In vitro and in vivo studies,” [0189] J. Natl. Cancer Inst., 60:379-384, 1978.
  • Boyle, et al., “Prevention of taxol induced neuropathy by glutamate,” [0190] Cancer Res., 37:290, 1996.
  • Cabral and Barlow, “Resistance to antimitotic agents as genetic probes of microtubule structure and function,” [0191] Pharmac. Ther., 52:159-171, 1991.
  • Cabral, Wible, Brenner, Brinkley, “Taxol-requiring mutant of Chinese hamster ovary cells with impaired mitotic spindle assembly,” [0192] J Cell Biol., 97:30-39, 1983.
  • Cortes, J. E. and Pazdur, R., “Docetaxel”, [0193] Journal of Clinical Oncology 13:2643-2655, 1995.
  • Deutsch et al., “Synthesis of congeners and prodrugs. 3. Water-soluble prodrugs of paclitaxel with potent antitumor activity,” [0194] J. Abed Chem., 32:788-792, 1989.
  • Duncan, et al., “Anticancer agents coupled to N-(2hydroxypropyl)methacryamide copolymers. 3. Evaluation of adriamycin conjugates against mouse leukemia L1210 in vivo,” [0195] J. Controlled Rel., 10:51-63, 1989.
  • Eiseman et al., “Plasma pharmacokinetics and tissue distribution of paclitaxel in CD2F1 mice,” [0196] Cancer Chemother. Pharmacol., 34:465-471, 1994.
  • Fidler, Gersten, Hart, “The biology of cancer invasion and metastasis,” [0197] Adv. Cancer Res., 28:149-250, 1987.
  • Foa, Norton, Seidman, “Taxol (paclitaxel): a novel anti-microtubule agent with remarkable anti-neoplastic activity,” [0198] Int. J Clin. Lab. Res., 24:6-14, 1994.
  • Goldspiel, “Taxol pharmaceutical issues: preparation, administration, stability, and compatibility with other medications,” [0199] Ann. Pharmacotherapy, 28:S23-26, 1994.
  • Greenwald et al., “Highly water soluble taxol derivative: 2′-polyethylene glycol esters as potential products”, [0200] Bioorganic & Medicinal Chemistry Letters, 4:2465-2470, 1994.
  • Greenwald et al., “Highly water soluble Taxol derivatives, 7-polyethylene glycol esters as potential products,” [0201] J. Org. Chem., 60:331-336, 1995.
  • Greenwald, et al., “Drug delivery systems: Water [0202] soluble taxol 2′-poly(ethylene glycol) ester prodrugs-design and in vivo effectiveness,” J. Med. Chem., 39:424-431, 1996.
  • Hirano et al., “Polymeric derivatives of activated cyclophosphamide as drug delivery systems in antitumor therapy”, [0203] Macromol. Chem., 180:1125-1130, 1979.
  • Hoes et al., “Optimization of macromolecular prodrugs of the antitumor antibiotic adriamycin”, [0204] J. Controlled Release, 2:205-213, 1985.
  • Holmes, Kudelka, Kavanagh, Huber, Ajani, Valero, “Current status of clinical trials with paclitaxel and docetaxel,” [0205] In: Taxane Anticancer Agents: Basic Science and Current Status, Georg, Chen, Ojima, Vyas, eds., American Chemical Society, Washington, D.C., 31-57, 1995.
  • Horwitz et al., “Taxol, mechanisms of action and resistance,” [0206] J. Natl. Cancer Inst. Monographs No. 15, pp. 55-61, 1993.
  • Kato, et al, “Antitumor activity of 1-barabinofuranosylcytosine conjugated with polyglutamic acid and its derivative,” [0207] Cancer Res., 44:25-30, 1984.
  • Kopecek and Kopeckova, “Targetable water-soluble polymeric anticancer drugs: achievements and unsolved problems,” [0208] Proceed Intern Symp. Control. Rel. Bioact. Mater., 20:190-191, 1993.
  • Kopecek, “The potential of water-soluble polymeric carriers in targeted and site-specific drug delivery”, [0209] J. Controlled Release, 11:279-290, 1990.
  • Li, et al., “Synthesis and evaluation of water-soluble polyethylene glycol paclitaxel conjugate as a paclitaxel prodrug,” [0210] Anti-Cancer Drugs, 7:642-648, 1996.
  • Liu, et al., “Evidence for involvement of tyrosine phosphorylation in taxol-induced apoptosis in a human ovarian tumor cell line,” [0211] Biochem. Pharmacol, 48:1265-1272, 1994.
  • Longnecker, et al., “High performance liquid chromatographic assay for Taxol in human plasma and urine and pharmacokinetics in a phase I trial,” [0212] Cancer Treat. Rep., 71:53-59, 1987.
  • Maeda and Matsumura, “Tumoritropic and lymphotropic principles of macromolecular drugs”, [0213] Critical Review in Therapeutic Drug Carrier Systems, 6:193-210, 1989.
  • Maeda, “SMANCS and polymer-conjugated macromolecular drugs: advantages in cancer chemotherapy,” [0214] Adv. Drug Delivery Rev., 6:181-193, 1991.
  • Maeda, Seymour, Miyamoto, “Conjugates of anticancer agents and polymers: Advantages of macromolecular therapeutics in vivo,” [0215] Bioconjug. Chem., 3:351-362, 1992.
  • Magri and Kingston, “Modified taxols. 2. Oxidation products of taxol,” [0216] 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,” [0217] J. Med. Chem., 35:145-151, 1992.
  • Milas, et al., “Kinetics of mitotic arrest and apoptosis in murine mammary and ovarian tumors treated with taxol,” [0218] Proc. Am. Assoc. Cancer Chemother. Pharmacol., 35:297-303, 1995.
  • Mosmann, T., “Rapid colormetric assay for cellular growth and survival: application to proliferation and cytotoxic assay,” [0219] J. Immunol. Methods, 65:55-63, 1983.
  • Nicolaou, Riemer, Kerr, Rideout, Wrasidio, “Design, synthesis and biological activity of protaxols,” [0220] Nature, 364:464-466, 1993.
  • Oliver, S. J. et al, Suppression of collagen-induced arthritis using an angiogenesis inhibitor, AGM-1470, and a microtubule stabilizer, Taxol.” [0221] Cellular Immunology 157:291-299, 1994.
  • Phillips-Hughes and Kandarpa, “Restenosis: pathophysiology and preventive strategies,” [0222] JVIR 7:321-333, 1996.
  • Reynolds, T., “Polymers help guide cancer drugs to tumor targets- and keep them there,” [0223] J. Natl. Cancer Institute, 87:1582-1584, 1995.
  • Rose, et al., “Preclinical antitumor activity of water-soluble paclitaxel derivatives,” [0224] Cancer Chemother. Pharmacol., 39:486-492, 1997.
  • Rowinsky and Donehower, “Review: Paclitaxel (Taxol),” [0225] N. Engl. J. Med., 332:1004-1014, 1995.
  • Rowinsky, Chaudhry, Cornblath, Donehower, “Phase I and pharmacologic study of paclitaxel and cisplatin with granulocyte colony-stimulating factor: neuromuscular toxicity is dose-limiting,” [0226] J. Clin. Oncol., 11:2010-2020, 1993.
  • 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,” [0227] Cancer Research, 48:4827-4833, 1988.
  • Serruys, De Jaegere, Kiemeneij et al., “A comparison of balloon-expandable-stent implantation with balloon angioplasty in patients with coronary artery disease,” N. [0228] Eng. J. Med., 331:489-495, 1994.
  • Sharma and Straubinger, “Novel taxol formulations: Preparation and Characterization of taxol-containing liposomes,” [0229] Pharm. Res. 11:889-896, 1994.
  • Trouet, et al., “A covalent linkage between daunorubicin and proteins that is stable in serum and reversible by lysosomal hydrolases, as required for a lysosomotropic drug-carrier conjugate: In vitro and in vivo studies,” [0230] Proc. Natl. Acad. Sci. U.S.A., 79:626-629, 1982.
  • U.S. Pat. No. 5,583,153 [0231]
  • U.S. Pat. No. 5,362,831 [0232]
  • van Heeswijk et al., “The synthesis and characterization of polypeptide-adriamycin conjugate and its complexes with adriamycin. [0233] Part 1”, J. Controlled Release, 1:301-315, 1985.
  • Vyas et al., “Phosphatase-activated prodrugs of paclitaxel,” [0234] In: Taxane Anticancer Agents: Basic Science and Current Status, Georg, Chen, Ojima, Vyas, eds., American Chemical Society, Washington, D.C., 124-137, 1995.
  • Weiss et al., “Hypersensitivity reactions from Taxol,” [0235] J. Clin. Oncol., 8:1263-1268, 1990.
  • Zhao, Z. and Kingston, D. G. I., “Modified taxols. 6. Preparation of water-soluble taxol phosphates,” [0236] J. Nat. Prod., 54:1607-1611, 1991.

Claims (51)

What is claimed is:
1. A composition comprising an anti-tumor drug selected from paclitaxel, docetaxel, etopside, teniposide, camptothecin or epothilone conjugated to polymer of water soluble amino acids.
2. The composition of
claim 1
wherein said 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 polymer is further defined as a copolymer of water soluble polyamino acids with polycaprolactone, polyglycolic acid, and polylactic acid and other polymers of poly(2-hydroxyethyl 1-glutamine), carboxymethyl dextran, hyaluronic acid, human serum albumin, polyalginic acid or a combination thereof.
5. The composition of
claim 1
, wherein said polymer has a molecular weight of from about 5,000 to about 100,000.
6. The composition of
claim 1
, wherein said polymer has a molecular weight of from about 20,000 to about 80,000.
7. The composition of
claim 1
, wherein said polymer has a molecular weight of about 25,000 to about 50,000.
8. The composition of
claim 1
, wherein said polymer comprises a water soluble polymer.
9. The composition of
claim 8
, wherein said water soluble polymer is conjugated to the 2′-and/or the 7-hydroxyl of paclitaxel or docetaxel.
10. The composition of
claim 8
, wherein said water soluble polymer is poly (l-glutamic acid), poly (d-glutamic acid), or poly (dl-glutamic acid).
11. The composition of
claim 8
, wherein said water soluble polymer is poly (l-aspartic acid), poly (d-aspartic acid), or poly (dl-aspartic acid).
12. The composition of
claim 1
, wherein said composition is dispersed in a pharmaceutically acceptable solution.
13. The composition of
claim 1
, wherein said composition comprises from about 2% to about 35% (w/w) anti-tumor drug.
14. The composition of
claim 10
, wherein said water soluble polymer contains 1 to 65 molecules of paclitaxel per chain.
15. The composition of
claim 14
, wherein said water soluble polymer contains 3 to 15 molecules of paclitaxel per chain.
16. A method of treating cancer in a subject comprising the steps of:
a) obtaining a composition comprising paclitaxel or docetaxel conjugated to water soluble polyamino acids and dispersed in a pharmaceutically acceptable solution;
b) administering said solution to said subject in an amount effective to treat said cancer wherein said treating cancer kills a cancer cell by mechanisms in addition to apoptosis.
17. The method of
claim 16
, wherein said composition comprises paclitaxel conjugated to poly-glutamic acids, poly-aspartic acids or poly-lysines.
18. The method of
claim 17
, wherein said composition comprises paclitaxel conjugated to poly-glutamic acids.
19. The method of
claim 16
, wherein said cancer is breast cancer, ovarian cancer, malignant melanoma, lung cancer, gastric cancer, prostate cancer, colon cancer, head and neck cancer, leukemia, or Kaposi's Sarcoma.
20. The method of
claim 19
, wherein said cancer is breast cancer.
21. The method of
claim 19
, wherein said cancer is ovarian cancer.
22. The method of
claim 19
, wherein said cancer is malignant melanoma.
23. The method of
claim 19
, wherein said cancer is lung cancer.
24. The method of
claim 19
, wherein said cancer is gastric cancer.
25. The method of
claim 19
, wherein said cancer is prostate cancer.
26. The method of
claim 19
, wherein said cancer is colon cancer.
27. The method of
claim 19
, wherein said cancer is head and neck cancer.
28. The method of
claim 19
, wherein said cancer is leukemia.
29. The method of
claim 19
, wherein said cancer is Kaposi's Sarcoma.
30. 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 water soluble polyamino acids.
31. The method of
claim 30
, wherein said composition comprises paclitaxel conjugated to poly-glutamic acids, poly-aspartic acids or poly-lysines.
32. The method of
claim 31
, wherein said composition comprises paclitaxel conjugated to poly-glutamic acid.
33. The method of
claim 30
, wherein said systemic autoimmune disease is rheumatoid arthritis.
34. 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 water soluble polyamino acids.
35. The method of
claim 34
, wherein said composition comprises paclitaxel conjugated to poly-glutamic acids, poly-aspartic acids or poly-lysines.
36. The method of
claim 35
, wherein said composition comprises paclitaxel conjugated to poly-glutamic acids.
37. The method of
claim 34
, wherein said subject is a coronary bypass, vascular surgery, organ transplant, coronary angioplasty, or arterial angioplasty patient.
38. The method of
claim 34
, wherein said composition is coated on a stent and said stent is implanted at the site of vascular trauma.
39. A pharmaceutical composition comprising paclitaxel conjugated to water soluble polyamino acids.
40. The pharmaceutical composition of
claim 39
, wherein said composition comprises paclitaxel conjugated to poly-glutamic acids, poly-aspartic acids or poly-lysines.
41. An implantable medical device, wherein said device is coated with a composition comprising paclitaxel conjugated to water soluble polyamino acids in an amount effective to inhibit smooth muscle cell proliferation.
42. The composition of
claim 41
, wherein said water soluble polyamino acids comprise poly (l-glutamic acid), poly (d-glutamic acid), or poly (dl-glutamic acid).
43. The implantable medical device of
claim 42
, further defined as a stent coated with said composition.
44. The implantable medical device of
claim 43
, wherein said stent is adapted to be used after balloon angioplasty and said composition is effective to inhibit restenosis.
45. A composition comprising water soluble polyamino acids conjugated to the 2′ or 7 hydroxyl of paclitaxel.
46. The composition of
claim 45
, wherein said composition comprises poly-glutamic acids conjugated to the 2′ or 7 hydroxyl of paclitaxel.
47. The composition of
claim 45
, wherein said composition comprises poly-aspartic acids conjugated to the 2′ or 7 hydroxyl of paclitaxel.
48. The composition of
claim 45
, wherein said composition comprises poly-lysines conjugated to the 2′ or 7 hydroxyl of paclitaxel.
49. The composition of
claim 46
, wherein said poly-glutamic acids are conjugated to the 2′ and the 7 hydroxyl of paclitaxel.
50. The composition of
claim 47
, wherein said poly-aspartic acids are conjugated to the 2′ and the 7 hydroxyl of paclitaxel.
51. The composition of
claim 48
, wherein said poly-lysines are conjugated to the 2′ and the 7 hydroxyl of paclitaxel.
US09/050,662 1996-03-12 1998-03-30 Water soluble paclitaxel derivatives Expired - Lifetime US6441025B2 (en)

Priority Applications (34)

Application Number Priority Date Filing Date Title
US09/050,662 US6441025B2 (en) 1996-03-12 1998-03-30 Water soluble paclitaxel derivatives
DK99916188T DK1028756T3 (en) 1998-03-30 1999-03-30 Water-soluble pacliataxel conjugates combined with radiation to treat cancer
DE69927350T DE69927350T2 (en) 1998-03-30 1999-03-30 WATER-SOLUBLE PACLITAXEL CONJUGATE COMBINED WITH IRRADIATION TO TREAT CANCER
TW093136237A TWI255183B (en) 1998-03-30 1999-03-30 Pharmaceutical compositions of water soluble camptothecin for the treatment of a tumor
AU34556/99A AU3455699A (en) 1998-03-30 1999-03-30 Water soluble paclitaxel derivatives
TW088104989A TWI246922B (en) 1998-03-30 1999-03-30 Pharmaceutical compositions of water soluble paclitaxel derivatives for the treatment of a tumor
AT99916188T ATE304867T1 (en) 1998-03-30 1999-03-30 WATER SOLUBLE PACLITAXEL CONJUGATES COMBINED WITH RADIATION FOR THE TREATMENT OF CANCER
MYPI99001207A MY121807A (en) 1998-03-30 1999-03-30 Water soluble paclitaxel derivatives
EP99916188A EP1028756B1 (en) 1998-03-30 1999-03-30 Water soluble paclitaxel conjugates combined with irradiation for the treatment of cancer
EP05007529A EP1598081A3 (en) 1998-03-30 1999-03-30 Conjugates of a water-soluble amino acid polymer and a drug
ES99916188T ES2249888T3 (en) 1998-03-30 1999-03-30 WATER SOLUBLE PACLITAXEL CONJUGATES, COMBINED WITH IRRADIATION FOR CANCER TREATMENT.
CA2303338A CA2303338C (en) 1998-03-30 1999-03-30 Water soluble paclitaxel derivatives
PCT/US1999/006870 WO1999049901A1 (en) 1998-03-30 1999-03-30 Water soluble paclitaxel derivatives
US10/146,809 US6884817B2 (en) 1996-03-12 2002-05-17 Water soluble paclitaxel derivatives
US10/153,818 US6515017B1 (en) 1998-03-30 2002-05-24 Water soluble paclitaxel derivatives
US10/243,045 US20030114518A1 (en) 1996-03-12 2002-09-12 Water soluble paclitaxel derivatives
US10/243,080 US20030114363A1 (en) 1996-03-12 2002-09-12 Water soluble paclitaxel derivatives
US10/243,046 US20030113335A1 (en) 1996-03-12 2002-09-12 Water soluble paclitaxel derivatives
US10/243,079 US20030114397A1 (en) 1996-03-12 2002-09-12 Water soluble paclitaxel derivatives
US10/282,490 US7135496B2 (en) 1996-03-12 2002-10-28 Water soluble paclitaxel derivatives
US10/282,570 US20030134793A1 (en) 1996-03-12 2002-10-28 Water soluble paclitaxel derivatives
US10/298,349 US20030130170A1 (en) 1996-03-12 2002-11-18 Water soluble paclitaxel derivatives
US10/298,327 US20030130178A1 (en) 1996-03-12 2002-11-18 Water soluble paclitaxel dervatives
US10/298,375 US20030130341A1 (en) 1996-03-12 2002-11-18 Water soluble paclitaxel derivatives
US10/300,031 US6730699B2 (en) 1998-03-30 2002-11-20 Water soluble paclitaxel derivatives
US10/310,331 US20030147807A1 (en) 1996-03-12 2002-12-05 Water soluble paclitaxel derivatives
US10/310,511 US20030124055A1 (en) 1996-03-12 2002-12-05 Water soluble paclitaxel derivatives
US10/354,431 US7060724B2 (en) 1996-03-12 2003-01-30 Water soluble paclitaxel derivatives
US10/620,238 US20040018960A1 (en) 1996-03-12 2003-07-14 Water soluble paclitaxel derivatives
US10/826,302 US20040198638A1 (en) 1998-03-30 2004-04-19 Water soluble paclitaxel derivatives
US11/253,810 US20060111273A1 (en) 1998-03-30 2005-10-20 Water soluble paclitaxel derivatives
US11/285,392 US20060135404A1 (en) 1996-03-12 2005-11-23 Water soluble paclitaxel derivatives
US11/513,390 US7384977B2 (en) 1996-03-12 2006-08-31 Water soluble paclitaxel prodrugs
US11/927,392 US20080153865A1 (en) 1998-03-30 2007-10-29 Water soluble paclitaxel derivatives

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US1318496P 1996-03-12 1996-03-12
US08/815,104 US5977163A (en) 1996-03-12 1997-03-11 Water soluble paclitaxel prodrugs
US09/050,662 US6441025B2 (en) 1996-03-12 1998-03-30 Water soluble paclitaxel derivatives

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US08/815,104 Continuation-In-Part US5977163A (en) 1996-03-12 1997-03-11 Water soluble paclitaxel prodrugs

Related Child Applications (6)

Application Number Title Priority Date Filing Date
PCT/US1999/006870 Continuation WO1999049901A1 (en) 1998-03-30 1999-03-30 Water soluble paclitaxel derivatives
PCT/US1999/006870 Continuation-In-Part WO1999049901A1 (en) 1998-03-30 1999-03-30 Water soluble paclitaxel derivatives
US09530601 Continuation 2001-01-11
US53060101A Continuation 1998-03-30 2001-01-11
US53060101A Continuation-In-Part 1998-03-30 2001-01-11
US10/146,809 Continuation US6884817B2 (en) 1996-03-12 2002-05-17 Water soluble paclitaxel derivatives

Publications (2)

Publication Number Publication Date
US20010034363A1 true US20010034363A1 (en) 2001-10-25
US6441025B2 US6441025B2 (en) 2002-08-27

Family

ID=21966621

Family Applications (22)

Application Number Title Priority Date Filing Date
US09/050,662 Expired - Lifetime US6441025B2 (en) 1996-03-12 1998-03-30 Water soluble paclitaxel derivatives
US10/146,809 Expired - Lifetime US6884817B2 (en) 1996-03-12 2002-05-17 Water soluble paclitaxel derivatives
US10/153,818 Expired - Lifetime US6515017B1 (en) 1998-03-30 2002-05-24 Water soluble paclitaxel derivatives
US10/243,046 Abandoned US20030113335A1 (en) 1996-03-12 2002-09-12 Water soluble paclitaxel derivatives
US10/243,079 Abandoned US20030114397A1 (en) 1996-03-12 2002-09-12 Water soluble paclitaxel derivatives
US10/243,080 Abandoned US20030114363A1 (en) 1996-03-12 2002-09-12 Water soluble paclitaxel derivatives
US10/243,045 Abandoned US20030114518A1 (en) 1996-03-12 2002-09-12 Water soluble paclitaxel derivatives
US10/282,490 Expired - Fee Related US7135496B2 (en) 1996-03-12 2002-10-28 Water soluble paclitaxel derivatives
US10/282,570 Abandoned US20030134793A1 (en) 1996-03-12 2002-10-28 Water soluble paclitaxel derivatives
US10/298,349 Abandoned US20030130170A1 (en) 1996-03-12 2002-11-18 Water soluble paclitaxel derivatives
US10/298,327 Abandoned US20030130178A1 (en) 1996-03-12 2002-11-18 Water soluble paclitaxel dervatives
US10/298,375 Abandoned US20030130341A1 (en) 1996-03-12 2002-11-18 Water soluble paclitaxel derivatives
US10/300,031 Expired - Lifetime US6730699B2 (en) 1998-03-30 2002-11-20 Water soluble paclitaxel derivatives
US10/310,331 Abandoned US20030147807A1 (en) 1996-03-12 2002-12-05 Water soluble paclitaxel derivatives
US10/310,511 Abandoned US20030124055A1 (en) 1996-03-12 2002-12-05 Water soluble paclitaxel derivatives
US10/354,431 Expired - Fee Related US7060724B2 (en) 1996-03-12 2003-01-30 Water soluble paclitaxel derivatives
US10/620,238 Abandoned US20040018960A1 (en) 1996-03-12 2003-07-14 Water soluble paclitaxel derivatives
US10/826,302 Abandoned US20040198638A1 (en) 1998-03-30 2004-04-19 Water soluble paclitaxel derivatives
US11/253,810 Abandoned US20060111273A1 (en) 1998-03-30 2005-10-20 Water soluble paclitaxel derivatives
US11/285,392 Abandoned US20060135404A1 (en) 1996-03-12 2005-11-23 Water soluble paclitaxel derivatives
US11/513,390 Expired - Fee Related US7384977B2 (en) 1996-03-12 2006-08-31 Water soluble paclitaxel prodrugs
US11/927,392 Abandoned US20080153865A1 (en) 1998-03-30 2007-10-29 Water soluble paclitaxel derivatives

Family Applications After (21)

Application Number Title Priority Date Filing Date
US10/146,809 Expired - Lifetime US6884817B2 (en) 1996-03-12 2002-05-17 Water soluble paclitaxel derivatives
US10/153,818 Expired - Lifetime US6515017B1 (en) 1998-03-30 2002-05-24 Water soluble paclitaxel derivatives
US10/243,046 Abandoned US20030113335A1 (en) 1996-03-12 2002-09-12 Water soluble paclitaxel derivatives
US10/243,079 Abandoned US20030114397A1 (en) 1996-03-12 2002-09-12 Water soluble paclitaxel derivatives
US10/243,080 Abandoned US20030114363A1 (en) 1996-03-12 2002-09-12 Water soluble paclitaxel derivatives
US10/243,045 Abandoned US20030114518A1 (en) 1996-03-12 2002-09-12 Water soluble paclitaxel derivatives
US10/282,490 Expired - Fee Related US7135496B2 (en) 1996-03-12 2002-10-28 Water soluble paclitaxel derivatives
US10/282,570 Abandoned US20030134793A1 (en) 1996-03-12 2002-10-28 Water soluble paclitaxel derivatives
US10/298,349 Abandoned US20030130170A1 (en) 1996-03-12 2002-11-18 Water soluble paclitaxel derivatives
US10/298,327 Abandoned US20030130178A1 (en) 1996-03-12 2002-11-18 Water soluble paclitaxel dervatives
US10/298,375 Abandoned US20030130341A1 (en) 1996-03-12 2002-11-18 Water soluble paclitaxel derivatives
US10/300,031 Expired - Lifetime US6730699B2 (en) 1998-03-30 2002-11-20 Water soluble paclitaxel derivatives
US10/310,331 Abandoned US20030147807A1 (en) 1996-03-12 2002-12-05 Water soluble paclitaxel derivatives
US10/310,511 Abandoned US20030124055A1 (en) 1996-03-12 2002-12-05 Water soluble paclitaxel derivatives
US10/354,431 Expired - Fee Related US7060724B2 (en) 1996-03-12 2003-01-30 Water soluble paclitaxel derivatives
US10/620,238 Abandoned US20040018960A1 (en) 1996-03-12 2003-07-14 Water soluble paclitaxel derivatives
US10/826,302 Abandoned US20040198638A1 (en) 1998-03-30 2004-04-19 Water soluble paclitaxel derivatives
US11/253,810 Abandoned US20060111273A1 (en) 1998-03-30 2005-10-20 Water soluble paclitaxel derivatives
US11/285,392 Abandoned US20060135404A1 (en) 1996-03-12 2005-11-23 Water soluble paclitaxel derivatives
US11/513,390 Expired - Fee Related US7384977B2 (en) 1996-03-12 2006-08-31 Water soluble paclitaxel prodrugs
US11/927,392 Abandoned US20080153865A1 (en) 1998-03-30 2007-10-29 Water soluble paclitaxel derivatives

Country Status (11)

Country Link
US (22) US6441025B2 (en)
EP (2) EP1028756B1 (en)
AT (1) ATE304867T1 (en)
AU (1) AU3455699A (en)
CA (1) CA2303338C (en)
DE (1) DE69927350T2 (en)
DK (1) DK1028756T3 (en)
ES (1) ES2249888T3 (en)
MY (1) MY121807A (en)
TW (2) TWI255183B (en)
WO (1) WO1999049901A1 (en)

Cited By (73)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20020197261A1 (en) * 2001-04-26 2002-12-26 Chun Li Therapeutic agent/ligand conjugate compositions, their methods of synthesis and use
US20030113335A1 (en) * 1996-03-12 2003-06-19 Pg-Txl Company, L.P. Water soluble paclitaxel derivatives
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
US20030211973A1 (en) * 2000-03-17 2003-11-13 Cell Therapeutics, Inc. Polyglutamic acid-camptothecin conjugates and methods of preparation
US20030232968A1 (en) * 2001-12-21 2003-12-18 Chun Li Dendritic poly (amino acid) carriers and methods of use
WO2004011034A1 (en) * 2002-07-31 2004-02-05 General Electric Company Conjugated lysine copolymers for drug delivery and imaging
US6764507B2 (en) 2000-10-16 2004-07-20 Conor Medsystems, Inc. Expandable medical device with improved spatial distribution
US20040143321A1 (en) * 2002-11-08 2004-07-22 Conor Medsystems, Inc. Expandable medical device and method for treating chronic total occlusions with local delivery of an angiogenic factor
US20040166058A1 (en) * 2002-11-07 2004-08-26 Board Of Regents, The University Of Texas System Ethylenedicysteine (EC)-drug conjugates, compositions and methods for tissue specific disease imaging
US20040185053A1 (en) * 2002-12-13 2004-09-23 Immunomedics, Inc. Immunoconjugates with an intracellularly-cleavable linkage
US20050079133A1 (en) * 1999-10-25 2005-04-14 Yang David J. Ethylenedicysteine (EC)-drug conjugates, compositions and methods for tissue specific disease imaging
US20050129619A1 (en) * 2003-12-10 2005-06-16 Board Of Regents, The University Of Texas System N2S2 chelate-targeting ligand conjugates
US20060002975A1 (en) * 2002-11-08 2006-01-05 Conor Medsystems, Inc. Method and apparatus for reducing tissue damage after ischemic injury
US20060193865A1 (en) * 2002-12-13 2006-08-31 Immunomedics, Inc. Camptothecin-binding moiety conjugates
US20060258736A1 (en) * 2005-05-12 2006-11-16 Bristol-Myers Squibb Company Dosing regimen
US20070020337A1 (en) * 2002-03-29 2007-01-25 Maurizio Zenoni Paclitaxel-based antitumor formulation
US20070092440A1 (en) * 2003-05-09 2007-04-26 Edgar Braendle Bone localising radiopharmaceutical and tubulin-interacting compound combinatorial
US20080107598A1 (en) * 2006-10-05 2008-05-08 Yang David J Efficient Synthesis of Chelators for Nuclear Imaging and Radiotherapy: Compositions and Applications
US7615208B2 (en) 1999-10-25 2009-11-10 Board Of Regents, The University Of Texas System Metal ion-labeled bis-aminoethanethiol-targeting ligand conjugates, compositions, and methods for tissue-specific disease imaging
US20100015046A1 (en) * 2002-12-13 2010-01-21 Immunomedics, Inc. Camptothecin-Binding Moiety Conjugates
US20100104589A1 (en) * 2002-12-13 2010-04-29 Immunomedics, Inc. Immunoconjugates with an Intracellularly-Cleavable Linkage
US7750041B2 (en) * 2001-03-26 2010-07-06 Bayer Schering Pharma Aktiengesellschaft Preparation for the prophylaxis of restenosis
US7811622B2 (en) 1995-06-07 2010-10-12 Cook Incorporated Coated implantable medical device
US7819912B2 (en) 1998-03-30 2010-10-26 Innovational Holdings Llc Expandable medical device with beneficial agent delivery mechanism
US7850727B2 (en) 2001-08-20 2010-12-14 Innovational Holdings, Llc Expandable medical device for delivery of beneficial agent
US7850728B2 (en) 2000-10-16 2010-12-14 Innovational Holdings Llc Expandable medical device for delivery of beneficial agent
US20110070156A1 (en) * 2002-06-14 2011-03-24 Immunomedics, Inc. Combining Radioimmunotherapy and Antibody-Drug Conjugates for Improved Cancer Therapy
US8182527B2 (en) 2001-05-07 2012-05-22 Cordis Corporation Heparin barrier coating for controlled drug release
US8197881B2 (en) 2003-09-22 2012-06-12 Conor Medsystems, Inc. Method and apparatus for loading a beneficial agent into an expandable medical device
CN102558270A (en) * 2012-03-09 2012-07-11 辽宁新中现代医药有限公司 20(S) and 20(R)-dammarane-3beta,12beta,20,25-tetrol derivative and preparation method and application thereof
US8236048B2 (en) 2000-05-12 2012-08-07 Cordis Corporation Drug/drug delivery systems for the prevention and treatment of vascular disease
US8257305B2 (en) 2002-09-20 2012-09-04 Bayer Pharma Aktiengesellschaft Medical device for dispensing medicaments
US8303609B2 (en) 2000-09-29 2012-11-06 Cordis Corporation Coated medical devices
US20120301481A1 (en) * 2005-07-08 2012-11-29 National Institutes Of Health Targeting poly-gamma-glutamic acid to treat staphylococcus epidermidis and related infections
US8349390B2 (en) 2002-09-20 2013-01-08 Conor Medsystems, Inc. Method and apparatus for loading a beneficial agent into an expandable medical device
US8361537B2 (en) 1998-03-30 2013-01-29 Innovational Holdings, Llc Expandable medical device with beneficial agent concentration gradient
US8366660B2 (en) 2006-11-20 2013-02-05 Lutonix, Inc. Drug releasing coatings for medical devices
US8366662B2 (en) 2006-11-20 2013-02-05 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
US8414910B2 (en) 2006-11-20 2013-04-09 Lutonix, Inc. Drug releasing coatings for medical devices
US8420086B2 (en) 2002-12-13 2013-04-16 Immunomedics, Inc. Camptothecin conjugates of anti-CD22 antibodies for treatment of B cell diseases
US8425459B2 (en) 2006-11-20 2013-04-23 Lutonix, Inc. Medical device rapid drug releasing coatings comprising a therapeutic agent and a contrast agent
US8430055B2 (en) 2008-08-29 2013-04-30 Lutonix, Inc. Methods and apparatuses for coating balloon catheters
US8449901B2 (en) 2003-03-28 2013-05-28 Innovational Holdings, Llc Implantable medical device with beneficial agent concentration gradient
US20130231292A1 (en) * 2012-03-02 2013-09-05 Wei Zhou Synergistic anti-proliferation activity of TAS-108 with mTOR inhibitors against cancer cells
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
US8998846B2 (en) 2006-11-20 2015-04-07 Lutonix, Inc. Drug releasing coatings for balloon catheters
US9028833B2 (en) 2012-12-13 2015-05-12 Immunomedics, Inc. Dosages of immunoconjugates of antibodies and SN-38 for improved efficacy and decreased toxicity
US9107960B2 (en) 2012-12-13 2015-08-18 Immunimedics, Inc. Antibody-SN-38 immunoconjugates with a CL2A linker
US20160106672A1 (en) * 2004-05-03 2016-04-21 Merrimack Pharmaceuticals, Inc. Liposomes Useful for Drug Delivery
US9382329B2 (en) 2012-08-14 2016-07-05 Ibc Pharmaceuticals, Inc. Disease therapy by inducing immune response to Trop-2 expressing cells
US9402935B2 (en) 2006-11-20 2016-08-02 Lutonix, Inc. Treatment of asthma and chronic obstructive pulmonary disease with anti-proliferate and anti-inflammatory drugs
US9492566B2 (en) 2012-12-13 2016-11-15 Immunomedics, Inc. Antibody-drug conjugates and uses thereof
US9700704B2 (en) 2006-11-20 2017-07-11 Lutonix, Inc. Drug releasing coatings for balloon catheters
US9707302B2 (en) 2013-07-23 2017-07-18 Immunomedics, Inc. Combining anti-HLA-DR or anti-Trop-2 antibodies with microtubule inhibitors, PARP inhibitors, bruton kinase inhibitors or phosphoinositide 3-kinase inhibitors significantly improves therapeutic outcome in cancer
US9737640B2 (en) 2006-11-20 2017-08-22 Lutonix, Inc. Drug releasing coatings for medical devices
US9737528B2 (en) 2004-05-03 2017-08-22 Ipsen Biopharm Ltd. Liposomes useful for drug delivery to the brain
US9770517B2 (en) 2002-03-01 2017-09-26 Immunomedics, Inc. Anti-Trop-2 antibody-drug conjugates and uses thereof
US9797907B2 (en) 2015-04-22 2017-10-24 Immunomedics, Inc. Isolation, detection, diagnosis and/or characterization of circulating Trop-2-positive cancer cells
US9931417B2 (en) 2012-12-13 2018-04-03 Immunomedics, Inc. Antibody-SN-38 immunoconjugates with a CL2A linker
US10058621B2 (en) 2015-06-25 2018-08-28 Immunomedics, Inc. Combination therapy with anti-HLA-DR antibodies and kinase inhibitors in hematopoietic cancers
US10137196B2 (en) 2012-12-13 2018-11-27 Immunomedics, Inc. Dosages of immunoconjugates of antibodies and SN-38 for improved efficacy and decreased toxicity
US10195175B2 (en) 2015-06-25 2019-02-05 Immunomedics, Inc. Synergistic effect of anti-Trop-2 antibody-drug conjugate in combination therapy for triple-negative breast cancer when used with microtubule inhibitors or PARP inhibitors
US10206918B2 (en) 2012-12-13 2019-02-19 Immunomedics, Inc. Efficacy of anti-HLA-DR antiboddy drug conjugate IMMU-140 (hL243-CL2A-SN-38) in HLA-DR positive cancers
US10266605B2 (en) 2016-04-27 2019-04-23 Immunomedics, Inc. Efficacy of anti-trop-2-SN-38 antibody drug conjugates for therapy of tumors relapsed/refractory to checkpoint inhibitors
US10413539B2 (en) 2012-12-13 2019-09-17 Immunomedics, Inc. Therapy for metastatic urothelial cancer with the antibody-drug conjugate, sacituzumab govitecan (IMMU-132)
US10456360B2 (en) 2015-10-16 2019-10-29 Ipsen Biopharm Ltd. Stabilizing camptothecin pharmaceutical compositions
US10744129B2 (en) 2012-12-13 2020-08-18 Immunomedics, Inc. Therapy of small-cell lung cancer (SCLC) with a topoisomerase-I inhibiting antibody-drug conjugate (ADC) targeting Trop-2
US10799597B2 (en) 2017-04-03 2020-10-13 Immunomedics, Inc. Subcutaneous administration of antibody-drug conjugates for cancer therapy
US10918734B2 (en) 2017-03-27 2021-02-16 Immunomedics, Inc. Treatment of high Trop-2 expressing triple negative breast cancer (TNBC) with sacituzumab govitecan (IMMU-132) overcomes homologous recombination repair (HRR) rescue mediated by Rad51
US10954305B2 (en) 2016-02-10 2021-03-23 Immunomedics, Inc. Combination of ABCG2 inhibitors with sacituzumab govitecan (IMMU-132) overcomes resistance to SN-38 in Trop-2 expressing cancers
US11253606B2 (en) 2013-07-23 2022-02-22 Immunomedics, Inc. Combining anti-HLA-DR or anti-Trop-2 antibodies with microtubule inhibitors, PARP inhibitors, Bruton kinase inhibitors or phosphoinositide 3-kinase inhibitors significantly improves therapeutic outcome in cancer

Families Citing this family (238)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5439686A (en) * 1993-02-22 1995-08-08 Vivorx Pharmaceuticals, Inc. Methods for in vivo delivery of substantially water insoluble pharmacologically active agents and compositions useful therefor
US8137684B2 (en) * 1996-10-01 2012-03-20 Abraxis Bioscience, Llc Formulations of pharmacological agents, methods for the preparation thereof and methods for the use thereof
US20050163818A1 (en) * 1996-11-05 2005-07-28 Hsing-Wen Sung Drug-eluting device chemically treated with genipin
US20050043376A1 (en) * 1996-12-03 2005-02-24 Danishefsky Samuel J. Synthesis of epothilones, intermediates thereto, analogues and uses thereof
DE69734362T2 (en) 1996-12-03 2006-07-20 Sloan-Kettering Institute For Cancer Research SYNTHESIS OF EPOTHILONES, INTERMEDIATE PRODUCTS, ANALOGUES AND USES THEREOF
US6867305B2 (en) 1996-12-03 2005-03-15 Sloan-Kettering Institute For Cancer Research Synthesis of epothilones, intermediates thereto and analogues thereof
US6204388B1 (en) * 1996-12-03 2001-03-20 Sloan-Kettering Institute For Cancer Research Synthesis of epothilones, intermediates thereto and analogues thereof
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
IL134419A0 (en) * 1997-08-09 2001-04-30 Schering Ag Epothilone derivatives, process for the preparation thereof and pharmaceutical compositions containing the same
US7713297B2 (en) * 1998-04-11 2010-05-11 Boston Scientific Scimed, Inc. Drug-releasing stent with ceramic-containing layer
US20020099438A1 (en) * 1998-04-15 2002-07-25 Furst Joseph G. Irradiated stent coating
US20030040790A1 (en) * 1998-04-15 2003-02-27 Furst Joseph G. Stent coating
US8070796B2 (en) 1998-07-27 2011-12-06 Icon Interventional Systems, Inc. Thrombosis inhibiting graft
US7967855B2 (en) * 1998-07-27 2011-06-28 Icon Interventional Systems, Inc. Coated medical device
CA2360952A1 (en) * 1999-02-11 2000-08-17 Schering Aktiengesellschaft Epothilon derivatives, method for the production and the use thereof as pharmaceuticals
US20020058286A1 (en) * 1999-02-24 2002-05-16 Danishefsky Samuel J. Synthesis of epothilones, intermediates thereto and analogues thereof
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
AU772750C (en) * 1999-04-30 2005-02-24 Schering Aktiengesellschaft 6-alkenyl-, 6-alkinyl- and 6-epoxy-epothilone derivatives, process for their production, and their use in pharmaceutical preparations
US7141234B1 (en) * 1999-09-21 2006-11-28 The United States Of America As Represented By The Department Of Health And Human Services Imaging of drug accumulation as a guide to antitumor therapy
CN1607962A (en) * 1999-10-12 2005-04-20 细胞治疗公司 Manufacture of polyglutamate-therapeutic agent conjugates
CA2388844A1 (en) * 1999-11-12 2001-05-25 Angiotech Pharmaceuticals, Inc. Compositions and methods for treating disease utilizing a combination of radioactive therapy and cell-cycle inhibitors
PL358335A1 (en) * 2000-03-17 2004-08-09 Cell Therapeutics, Inc. Polyglutamic acid-camptothecin conjugates and methods of preparation
GB0011903D0 (en) * 2000-05-18 2000-07-05 Astrazeneca Ab Combination chemotherapy
AU7007001A (en) 2000-06-22 2002-01-02 Nitromed Inc Nitrosated and nitrosylated taxanes, compositions and methods of use
EP2080511A3 (en) * 2000-08-22 2009-09-30 Shire LLC Active agent delivery systems and methods for protecting and administering active agents
JP4583756B2 (en) * 2000-10-31 2010-11-17 クック インコーポレイテッド Medical instruments
EP1401374B1 (en) * 2000-11-14 2010-03-31 Shire LLC A novel pharmaceutical compound containing atenolol and methods of making and using same
US8394813B2 (en) 2000-11-14 2013-03-12 Shire Llc Active agent delivery systems and methods for protecting and administering active agents
AU2001298033B2 (en) * 2000-11-14 2007-08-09 Shire Llc A novel pharmaceutical compound containing abacavir sulfate and methods of making and using same
WO2002058699A1 (en) * 2001-01-25 2002-08-01 Bristol-Myers Squibb Company Pharmaceutical forms of epothilones for oral administration
RU2003126171A (en) * 2001-01-25 2005-02-27 Бристол-Маерс Сквибб Компани (Us) Parenteral composition containing analogs of epothilone
US20030108585A1 (en) * 2001-05-04 2003-06-12 Roe R. Michael Polymer conjugates of insecticidal peptides or nucleic acids or insecticides and methods of use thereof
US7727221B2 (en) 2001-06-27 2010-06-01 Cardiac Pacemakers Inc. Method and device for electrochemical formation of therapeutic species in vivo
US7169752B2 (en) * 2003-09-30 2007-01-30 New River Pharmaceuticals Inc. Compounds and compositions for prevention of overdose of oxycodone
US20050025808A1 (en) * 2001-09-24 2005-02-03 Herrmann Robert A. Medical devices and methods for inhibiting smooth muscle cell proliferation
ES2625340T3 (en) * 2001-10-15 2017-07-19 Crititech, Inc. Compositions and methods for the administration of poorly water soluble drugs and treatment methods
US8016881B2 (en) 2002-07-31 2011-09-13 Icon Interventional Systems, Inc. Sutures and surgical staples for anastamoses, wound closures, and surgical closures
US7649006B2 (en) * 2002-08-23 2010-01-19 Sloan-Kettering Institute For Cancer Research Synthesis of epothilones, intermediates thereto and analogues thereof
DK1767535T3 (en) 2002-08-23 2010-04-12 Sloan Kettering Inst Cancer Synthesis of epothilones, their intermediates, analogs and their use
US6921769B2 (en) 2002-08-23 2005-07-26 Sloan-Kettering Institute For Cancer Research Synthesis of epothilones, intermediates thereto and analogues thereof
MXPA05002444A (en) 2002-09-06 2005-09-30 Insert Therapeutics Inc Cyclodextrin-based polymers for delivering the therapeutic agents covalently bound thereto.
US20040047835A1 (en) * 2002-09-06 2004-03-11 Cell Therapeutics, Inc. Combinatorial drug therapy using polymer drug conjugates
SI1585548T1 (en) 2002-12-09 2018-11-30 Abraxis Bioscience, Llc Compositions and methods of delivery of pharmacological agents
GB0305928D0 (en) * 2003-03-14 2003-04-23 Novartis Ag Organic compounds
EP1475105A1 (en) * 2003-05-09 2004-11-10 Schering AG Bone localising radiopharmaceutical and tubulin-interacting compound combinatorial radiotherapy
US7344491B1 (en) 2003-11-26 2008-03-18 Nanobiomagnetics, Inc. Method and apparatus for improving hearing
US8651113B2 (en) * 2003-06-18 2014-02-18 Swr&D Inc. Magnetically responsive nanoparticle therapeutic constructs and methods of making and using
US7723311B2 (en) * 2003-06-18 2010-05-25 Nanobiomagnetics, Inc. Delivery of bioactive substances to target cells
US7229979B2 (en) * 2003-06-23 2007-06-12 Immune Modulation, Inc. Hypoestoxides, derivatives and agonists thereof for use as stent-coating agents
US20050152979A1 (en) * 2003-09-05 2005-07-14 Cell Therapeutics, Inc. Hydrophobic drug compositions containing reconstitution enhancer
US20050148534A1 (en) * 2003-09-22 2005-07-07 Castellino Angelo J. Small molecule compositions and methods for increasing drug efficiency using compositions thereof
WO2005079861A2 (en) * 2004-02-13 2005-09-01 Safeway Investments Ltd. Polymeric water soluble prodrugs
US7317070B1 (en) 2004-03-12 2008-01-08 Sigma-Aldrich Co. Process for the preparation of polyamino acids
WO2006138673A2 (en) 2005-06-15 2006-12-28 Cardiome Pharma Corp. Synthetic processes for the preparation of aminocyclohexyl ether compounds
US7977373B2 (en) 2004-04-01 2011-07-12 Cardiome Pharma Corp. Prodrugs of ion channel modulating compounds and uses thereof
CA2561733A1 (en) * 2004-04-01 2005-10-13 Cardiome Pharma Corp. Pegylated ion channel modulating compounds
US7705036B2 (en) * 2004-04-01 2010-04-27 Cardiome Pharma Corp. Deuterated aminocyclohexyl ether compounds and processes for preparing same
EP1802648A2 (en) * 2004-09-10 2007-07-04 Cornell Research Foundation, Inc. Betulinol derivatives as anti-cancer agents
WO2006035340A1 (en) * 2004-09-27 2006-04-06 Koninklijke Philips Electronics N.V. Method for selecting the sensitivity of an input device
WO2006042146A2 (en) * 2004-10-07 2006-04-20 Emory University Multifunctional nanoparticles conjugates and their use
US20050148669A1 (en) * 2004-10-21 2005-07-07 Daniel Amato Amino acid esters as nutrient supplements and methods of use
CN100361985C (en) * 2004-10-26 2008-01-16 中国科学院长春应用化学研究所 Paclitaxol predrug of biodegradable polymer and its synthesis method
JP5159314B2 (en) 2004-11-18 2013-03-06 カーディオム ファーマ コーポレイション Method for synthesizing aminocyclohexyl ether compound
KR20070095916A (en) * 2004-11-26 2007-10-01 스텐토믹스 인코포레이티드 Chelating and binding chemicals to a medical implant
US20120269886A1 (en) 2004-12-22 2012-10-25 Nitto Denko Corporation Therapeutic agent for pulmonary fibrosis
CA2820728C (en) 2004-12-22 2016-01-26 Nitto Denko Corporation Drug carrier and drug carrier kit for inhibiting fibrosis
US8377918B2 (en) * 2005-01-31 2013-02-19 ACC Therapeutics Inc Apigenin for chemoprevention, and chemotherapy combined with therapeutic reagents
US7850645B2 (en) * 2005-02-11 2010-12-14 Boston Scientific Scimed, Inc. Internal medical devices for delivery of therapeutic agent in conjunction with a source of electrical power
DK2301531T3 (en) 2005-02-18 2018-07-30 Abraxis Bioscience Llc COMBINATIONS AND WAYS FOR ADMINISTRATING THERAPEUTIC SUBSTANCES AND COMBINATION THERAPY
US20060264914A1 (en) * 2005-03-03 2006-11-23 Icon Medical Corp. Metal alloys for medical devices
US8323333B2 (en) * 2005-03-03 2012-12-04 Icon Medical Corp. Fragile structure protective coating
US9107899B2 (en) 2005-03-03 2015-08-18 Icon Medical Corporation Metal alloys for medical devices
WO2006096251A2 (en) 2005-03-03 2006-09-14 Icon Medical Corp. Improved metal alloys for medical device
WO2006110197A2 (en) * 2005-03-03 2006-10-19 Icon Medical Corp. Polymer biodegradable medical device
US20060201601A1 (en) * 2005-03-03 2006-09-14 Icon Interventional Systems, Inc. Flexible markers
US7540995B2 (en) * 2005-03-03 2009-06-02 Icon Medical Corp. Process for forming an improved metal alloy stent
US7452501B2 (en) * 2005-03-03 2008-11-18 Icon Medical Corp. Metal alloy for a stent
US20060200048A1 (en) * 2005-03-03 2006-09-07 Icon Medical Corp. Removable sheath for device protection
US9820986B2 (en) * 2005-03-04 2017-11-21 Taiwan Hopaz Chems, Mfg. Co., Ltd. Glycopeptide compositions
JP4954983B2 (en) 2005-05-18 2012-06-20 ファーマサイエンス・インコーポレイテッド BIR domain binding compound
NO345390B1 (en) * 2005-08-31 2021-01-11 Abraxis Bioscience Llc Compositions comprising poorly water-soluble pharmaceuticals and antimicrobials
US8034765B2 (en) * 2005-08-31 2011-10-11 Abraxis Bioscience, Llc Compositions and methods for preparation of poorly water soluble drugs with increased stability
US7855279B2 (en) 2005-09-27 2010-12-21 Amunix Operating, Inc. Unstructured recombinant polymers and uses thereof
US7846445B2 (en) 2005-09-27 2010-12-07 Amunix Operating, Inc. Methods for production of unstructured recombinant polymers and uses thereof
JP2009509535A (en) * 2005-09-27 2009-03-12 アムニクス, インコーポレイテッド Proteinaceous drugs and their use
US9839667B2 (en) 2005-10-14 2017-12-12 Allergan, Inc. Prevention and treatment of ocular side effects with a cyclosporin
US20070110786A1 (en) * 2005-11-15 2007-05-17 Boston Scientific Scimed, Inc. Medical articles having enhanced therapeutic agent binding
ATE544802T1 (en) * 2005-12-05 2012-02-15 Nitto Denko Corp POLYGLUTAMAT-AMINO ACID CONJUGATES AND METHODS
EP1957065A1 (en) * 2005-12-06 2008-08-20 Cell Therapeutics, Inc. Estrogen cancer therapy
WO2010009146A1 (en) * 2008-07-15 2010-01-21 University Of Kansas Nanoclusters for delivery of poorly water soluble drug nanoparticles
US8906392B2 (en) * 2005-12-16 2014-12-09 University Of Kansas Nanocluster compositions and methods
US9572886B2 (en) 2005-12-22 2017-02-21 Nitto Denko Corporation Agent for treating myelofibrosis
US20070148697A1 (en) * 2005-12-27 2007-06-28 Boston Scientific Scimed, Inc. Methods and system for high throughput screening of polymer materials for medical devices
US8834912B2 (en) * 2005-12-30 2014-09-16 Boston Scientific Scimed, Inc. Medical devices having multiple charged layers
US20070154466A1 (en) * 2005-12-30 2007-07-05 Jan Weber Internal medical devices containing peroxide-converting catalysts
US8840660B2 (en) 2006-01-05 2014-09-23 Boston Scientific Scimed, Inc. Bioerodible endoprostheses and methods of making the same
AR054215A1 (en) 2006-01-20 2007-06-13 Eriochem Sa A PHARMACEUTICAL FORMULATION OF A TAXANE, A SOLID COMPOSITION OF A LIOFILIZED TAXAN FROM AN ACETIC ACID SOLUTION, A PROCEDURE FOR THE PREPARATION OF A SOLID COMPOSITION OF A TAXANE, A SOLUBILIZING COMPOSITION OF A LIOFILIZED TAXANE AND AN ELEMENTARY KIT
CN100431610C (en) * 2006-01-24 2008-11-12 中国科学院长春应用化学研究所 Amphiphilic tri-block copolymer-paclitaxel bonded drug and preparation method thereof
US8440214B2 (en) * 2006-01-31 2013-05-14 Boston Scientific Scimed, Inc. Medical devices for therapeutic agent delivery with polymeric regions that contain copolymers having both soft segments and uniform length hard segments
US8089029B2 (en) 2006-02-01 2012-01-03 Boston Scientific Scimed, Inc. Bioabsorbable metal medical device and method of manufacture
US20070224235A1 (en) 2006-03-24 2007-09-27 Barron Tenney Medical devices having nanoporous coatings for controlled therapeutic agent delivery
US20070225800A1 (en) * 2006-03-24 2007-09-27 Sahatjian Ronald A Methods and devices having electrically actuatable surfaces
US8187620B2 (en) 2006-03-27 2012-05-29 Boston Scientific Scimed, Inc. Medical devices comprising a porous metal oxide or metal material and a polymer coating for delivering therapeutic agents
US8048150B2 (en) 2006-04-12 2011-11-01 Boston Scientific Scimed, Inc. Endoprosthesis having a fiber meshwork disposed thereon
CA2652455C (en) 2006-05-16 2017-06-13 Aegera Therapeutics Inc. Iap bir domain binding compounds
KR100773029B1 (en) * 2006-06-16 2007-11-05 이화여자대학교 산학협력단 Water soluble micelle-forming and biodegradable cyclotriphosphazene-paclitaxol conjugate anticancer agent and the preparation method thereof
US8815275B2 (en) 2006-06-28 2014-08-26 Boston Scientific Scimed, Inc. Coatings for medical devices comprising a therapeutic agent and a metallic material
WO2008002778A2 (en) 2006-06-29 2008-01-03 Boston Scientific Limited Medical devices with selective coating
US8052743B2 (en) * 2006-08-02 2011-11-08 Boston Scientific Scimed, Inc. Endoprosthesis with three-dimensional disintegration control
JP2010503469A (en) 2006-09-14 2010-02-04 ボストン サイエンティフィック リミテッド Medical device having drug-eluting film
US8128689B2 (en) 2006-09-15 2012-03-06 Boston Scientific Scimed, Inc. Bioerodible endoprosthesis with biostable inorganic layers
CA2663303A1 (en) 2006-09-15 2008-03-20 Boston Scientific Limited Endoprosthesis with adjustable surface features
JP2010503489A (en) 2006-09-15 2010-02-04 ボストン サイエンティフィック リミテッド Biodegradable endoprosthesis and method for producing the same
CA2663271A1 (en) 2006-09-15 2008-03-20 Boston Scientific Limited Bioerodible endoprostheses and methods of making the same
CA2663220A1 (en) 2006-09-15 2008-03-20 Boston Scientific Limited Medical devices and methods of making the same
WO2008036548A2 (en) 2006-09-18 2008-03-27 Boston Scientific Limited Endoprostheses
US20080071349A1 (en) * 2006-09-18 2008-03-20 Boston Scientific Scimed, Inc. Medical Devices
EP2724722B1 (en) * 2006-10-20 2016-12-07 NeurEndo Pharma, LLC Method of restoring the incretin effect
US8168662B1 (en) 2006-11-06 2012-05-01 Poniard Pharmaceuticals, Inc. Use of picoplatin to treat colorectal cancer
US8168661B2 (en) * 2006-11-06 2012-05-01 Poniard Pharmaceuticals, Inc. Use of picoplatin to treat colorectal cancer
US8178564B2 (en) * 2006-11-06 2012-05-15 Poniard Pharmaceuticals, Inc. Use of picoplatin to treat colorectal cancer
US8173686B2 (en) 2006-11-06 2012-05-08 Poniard Pharmaceuticals, Inc. Use of picoplatin to treat colorectal cancer
US7981150B2 (en) 2006-11-09 2011-07-19 Boston Scientific Scimed, Inc. Endoprosthesis with coatings
EP2125065B1 (en) 2006-12-28 2010-11-17 Boston Scientific Limited Bioerodible endoprostheses and methods of making same
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
WO2008089258A2 (en) * 2007-01-16 2008-07-24 Cappella, Inc. Drug eluting medical device using polymeric therapeutics
US20080175881A1 (en) * 2007-01-18 2008-07-24 Boston Scientific Scimed, Inc. Blood-contacting medical devices for the release of nitric oxide and anti-restenotic agents
US20080176958A1 (en) 2007-01-24 2008-07-24 Insert Therapeutics, Inc. Cyclodextrin-based polymers for therapeutics delivery
US20080181852A1 (en) * 2007-01-29 2008-07-31 Nitto Denko Corporation Multi-functional Drug Carriers
US20110033528A1 (en) * 2009-08-05 2011-02-10 Poniard Pharmaceuticals, Inc. Stabilized picoplatin oral dosage form
US8070797B2 (en) 2007-03-01 2011-12-06 Boston Scientific Scimed, Inc. Medical device with a porous surface for delivery of a therapeutic agent
US8431149B2 (en) 2007-03-01 2013-04-30 Boston Scientific Scimed, Inc. Coated medical devices for abluminal drug delivery
CN101715342A (en) * 2007-03-19 2010-05-26 因弗拉布洛克制药公司 cobalamin taxane bioconjugates
TWI407971B (en) 2007-03-30 2013-09-11 Nitto Denko Corp Cancer cells and tumor-related fibroblasts
US8067054B2 (en) 2007-04-05 2011-11-29 Boston Scientific Scimed, Inc. Stents with ceramic drug reservoir layer and methods of making and using the same
WO2008124735A2 (en) * 2007-04-10 2008-10-16 Nitto Denko Corporation Multi-functional polyglutamate drug carriers
EP2155253A2 (en) * 2007-05-09 2010-02-24 Nitto Denko Corporation Polyglutamate conjugates and polyglutamate-amino acid conjugates having a plurality of drugs
WO2008141107A2 (en) * 2007-05-09 2008-11-20 Nitto Denko Corporation Compositions that include a hydrophobic compound and a polyamino acid conjugate
JP2010528122A (en) * 2007-05-09 2010-08-19 日東電工株式会社 Polymer combined with platinum drug
US7976915B2 (en) 2007-05-23 2011-07-12 Boston Scientific Scimed, Inc. Endoprosthesis with select ceramic morphology
US20100260832A1 (en) * 2007-06-27 2010-10-14 Poniard Pharmaceuticals, Inc. Combination therapy for ovarian cancer
TW200916094A (en) * 2007-06-27 2009-04-16 Poniard Pharmaceuticals Inc Stabilized picoplatin dosage form
US7942926B2 (en) 2007-07-11 2011-05-17 Boston Scientific Scimed, Inc. Endoprosthesis coating
US8002823B2 (en) 2007-07-11 2011-08-23 Boston Scientific Scimed, Inc. Endoprosthesis coating
CA2693057A1 (en) * 2007-07-16 2009-01-22 Poniard Pharmaceuticals, Inc. Oral formulations for picoplatin
EP2187988B1 (en) 2007-07-19 2013-08-21 Boston Scientific Limited Endoprosthesis having a non-fouling surface
US8815273B2 (en) 2007-07-27 2014-08-26 Boston Scientific Scimed, Inc. Drug eluting medical devices having porous layers
US7931683B2 (en) * 2007-07-27 2011-04-26 Boston Scientific Scimed, Inc. Articles having ceramic coated surfaces
US8221822B2 (en) 2007-07-31 2012-07-17 Boston Scientific Scimed, Inc. Medical device coating by laser cladding
JP2010535541A (en) 2007-08-03 2010-11-25 ボストン サイエンティフィック リミテッド Coating for medical devices with large surface area
WO2009023270A2 (en) 2007-08-15 2009-02-19 Amunix, Inc. Compositions and methods for modifying properties of biologically active polypeptides
US8052745B2 (en) 2007-09-13 2011-11-08 Boston Scientific Scimed, Inc. Endoprosthesis
EP2207570A2 (en) 2007-09-14 2010-07-21 Nitto Denko Corporation Drug carriers
AR063111A1 (en) 2007-10-03 2008-12-30 Eriochem Sa A PHARMACEUTICAL FORMULATION OF TAXANO
CA2700904C (en) * 2007-10-16 2014-10-14 Bionumerik Pharmaceuticals, Inc. C10-substituted camptothecin analogs
US8029554B2 (en) 2007-11-02 2011-10-04 Boston Scientific Scimed, Inc. Stent with embedded material
US7938855B2 (en) * 2007-11-02 2011-05-10 Boston Scientific Scimed, Inc. Deformable underlayer for stent
US8216632B2 (en) 2007-11-02 2012-07-10 Boston Scientific Scimed, Inc. Endoprosthesis coating
WO2009076272A2 (en) * 2007-12-07 2009-06-18 Boston Scientific Scimed, Inc. Drug coated stent with endosome-disrupting conjugate
CA2715348A1 (en) * 2008-02-08 2009-08-13 Poniard Pharmaceuticals, Inc. Use of picoplatin and bevacizumab to treat colorectal cancer
JP2011513412A (en) * 2008-03-06 2011-04-28 日東電工株式会社 Pharmaceutical composition for treating cancer comprising a polymer paclitaxel conjugate
EP2271380B1 (en) 2008-04-22 2013-03-20 Boston Scientific Scimed, Inc. Medical devices having a coating of inorganic material
US8932346B2 (en) 2008-04-24 2015-01-13 Boston Scientific Scimed, Inc. Medical devices having inorganic particle layers
US7998192B2 (en) 2008-05-09 2011-08-16 Boston Scientific Scimed, Inc. Endoprostheses
US8586019B2 (en) 2008-05-22 2013-11-19 Ramot At Tel-Aviv University Ltd. Conjugates of polymers having a therapeutically active agent and an angiogenesis targeting moiety attached thereto and uses thereof in the treatment of angiogenesis related diseases
US8658149B2 (en) * 2008-05-22 2014-02-25 Ramot At Tel-Aviv University Ltd. Conjugates of a polymer, a bisphosphonate and an anti-angiogenesis agent and uses thereof in the treatment and monitoring of bone related diseases
WO2009141827A2 (en) 2008-05-22 2009-11-26 Ramot At Tel Aviv University Ltd. A conjugate of a polymer, an anti-angiogenesis agent and a targeting moiety, and uses thereof in the treatment of bone related angiogenesis conditions
US20090297581A1 (en) * 2008-05-28 2009-12-03 Boston Scientific Scimed, Inc. Medical devices having electrodeposited coatings
US8236046B2 (en) * 2008-06-10 2012-08-07 Boston Scientific Scimed, Inc. Bioerodible endoprosthesis
US8449603B2 (en) 2008-06-18 2013-05-28 Boston Scientific Scimed, Inc. Endoprosthesis coating
EP2307068A2 (en) * 2008-06-25 2011-04-13 Boston Scientific Scimed, Inc. Medical devices having surface coatings
US8114148B2 (en) * 2008-06-25 2012-02-14 Boston Scientific Scimed, Inc. Medical devices for delivery of therapeutic agent in conjunction with galvanic corrosion
EP2310065A2 (en) * 2008-06-26 2011-04-20 Boston Scientific Scimed, Inc. Medical device coatings containing charged materials
CA2732412C (en) * 2008-07-30 2014-12-09 Lei Yu Retinoid-targeted drug carriers
US7985252B2 (en) 2008-07-30 2011-07-26 Boston Scientific Scimed, Inc. Bioerodible endoprosthesis
EP2307448B1 (en) * 2008-08-07 2015-04-22 Sigma-Aldrich Co. LLC Preparation of low molecular weight polylysine and polyornithine in high yield
EP2323709A2 (en) 2008-08-14 2011-05-25 Boston Scientific Scimed, Inc. Medical devices having electrodeposited conductive polymer coatings
US8382824B2 (en) 2008-10-03 2013-02-26 Boston Scientific Scimed, Inc. Medical implant having NANO-crystal grains with barrier layers of metal nitrides or fluorides
US8231980B2 (en) 2008-12-03 2012-07-31 Boston Scientific Scimed, Inc. Medical implants including iridium oxide
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
US8703717B2 (en) 2009-02-03 2014-04-22 Amunix Operating Inc. Growth hormone polypeptides and methods of making and using same
WO2010091122A1 (en) 2009-02-03 2010-08-12 Amunix, Inc. Extended recombinant polypeptides and compositions comprising same
US8267992B2 (en) 2009-03-02 2012-09-18 Boston Scientific Scimed, Inc. Self-buffering medical implants
US8071156B2 (en) 2009-03-04 2011-12-06 Boston Scientific Scimed, Inc. Endoprostheses
US8287937B2 (en) 2009-04-24 2012-10-16 Boston Scientific Scimed, Inc. Endoprosthese
US9849188B2 (en) 2009-06-08 2017-12-26 Amunix Operating Inc. Growth hormone polypeptides and methods of making and using same
EP2440228B8 (en) 2009-06-08 2023-02-22 Amunix Operating Inc. Glucose-regulating polypeptides and methods of making and using same
AU2010290131C1 (en) 2009-08-24 2015-12-03 Amunix Operating Inc. Coagulation factor VII compositions and methods of making and using same
US20120225825A1 (en) * 2009-11-23 2012-09-06 Cerulean Pharma Inc. Cyclodextrin-based polymers for therapeutic delivery
EP2519269A1 (en) * 2009-12-30 2012-11-07 Boston Scientific Scimed, Inc. Drug-delivery balloons
US20120309691A1 (en) * 2010-02-04 2012-12-06 Dapeng Zhou Tumor targeted delivery of immunomodulators by nanopolymers
US8702682B2 (en) * 2010-02-05 2014-04-22 Boston Scientific Scimed, Inc. Medical devices employing piezoelectric materials for delivery of therapeutic agents
KR20120140658A (en) 2010-02-12 2012-12-31 파마사이언스 인크. Iap bir domain binding compounds
US8398916B2 (en) * 2010-03-04 2013-03-19 Icon Medical Corp. Method for forming a tubular medical device
AR093275A1 (en) 2010-03-17 2015-05-27 Centro De Excelencia En Productos Y Procesos De Cordoba (Ceprocor) A PHARMACEUTICAL COMPOSITION SOLUBLE IN WATER THAT INCLUDES AT LEAST A THERAPEUTICALLY ACTIVE SUBSTANCE OF HYDROPHOBIC CHARACTERISTICS AND AT LEAST A SELECTED COMPOSITE BETWEEN THE SYMPHOGLIOPHYMPHYLOPHYLOPHYLOPHYLOPHYLOPHYLOPHYLOPHYLOPHYLOPHYLOPHYLOPHYL SOPHYLOPHYLOPHYLOPHYLOSPHYLOPHYLOPHYLOPHYL SINGLE WATER SOLUBLE COMPOSITION IN A WATER SOLUBLE PHARMACEUTICAL COMPOSITION THAT INCLUDES AT LEAST A THERAPEUTICALLY ACTIVE SUBSTANCE OF HYDROPHOBIC CHARACTERISTICS AND AT LEAST A SELECTED COMPOSITE
WO2011119573A1 (en) 2010-03-23 2011-09-29 Boston Scientific Scimed, Inc. Surface treated bioerodible metal endoprostheses
EP2552439B1 (en) 2010-03-29 2022-07-20 Abraxis BioScience, LLC Methods of enhancing drug delivery and effectiveness of therapeutic agents
US9393318B2 (en) 2010-03-29 2016-07-19 Abraxis Bioscience, Llc Methods of treating cancer
WO2011123830A2 (en) 2010-04-02 2011-10-06 Amunix Operating Inc. Alpha 1-antitrypsin compositions and methods of making and using same
DE102010022588A1 (en) 2010-05-27 2011-12-01 Hemoteq Ag Balloon catheter with a particle-free drug-releasing coating
MX347225B (en) 2010-06-04 2017-04-19 Abraxis Bioscience Llc * Methods of treatment of pancreatic cancer.
US9283279B2 (en) 2011-05-11 2016-03-15 Ramot At Tel-Aviv University Ltd. Targeted polymeric conjugates and uses thereof
WO2013090616A1 (en) 2011-12-13 2013-06-20 Io Therapeutics, Inc. Autoimmune disorder treatment using rxr agonists
US9532972B2 (en) 2012-02-07 2017-01-03 University Of Central Florida Research Foundation, Inc. Increasing taxane sensitivity in cancer cells
EP3549953A1 (en) 2012-02-15 2019-10-09 Bioverativ Therapeutics Inc. Recombinant factor viii proteins
PT2814840T (en) 2012-02-15 2020-01-28 Bioverativ Therapeutics Inc Factor viii compositions and methods of making and using same
JP6355563B2 (en) 2012-02-27 2018-07-11 アムニクス オペレーティング インコーポレイテッド XTEN conjugate composition and method for producing the same
EP2822598A4 (en) 2012-03-05 2016-04-13 Univ Ramot Polymers having therapeutically active agents conjugated thereto, processes of preparing same and uses thereof
WO2013146376A1 (en) 2012-03-27 2013-10-03 テルモ株式会社 Coating composition and medical device
US20150065443A1 (en) * 2012-04-06 2015-03-05 University of Pittsburgh - of the Commonwealth System of Higher Educatioon Embelin-based delivery system for water-insoluble active agents
RU2493848C1 (en) * 2012-06-14 2013-09-27 Геннадий Петрович Власов Biodegradable polymer carrier for anti-cancer drug delivery (versions)
GB201215289D0 (en) * 2012-08-28 2012-10-10 Medical Res Council Nanoparticle formulation
WO2014055493A1 (en) 2012-10-02 2014-04-10 Cerulean Pharma Inc. Methods and systems for polymer precipitation and generation of particles
CN103808852A (en) * 2012-11-15 2014-05-21 刘胜远 Thin-layer chromatography detection method of taxol in taxus chinensis
CN103641925B (en) * 2012-11-27 2016-08-17 王晖 Water solublity polysaccharide and the covalency polyacetylene compound of bearing taxanes, its preparation method and medical usage
TW202003554A (en) 2013-08-14 2020-01-16 美商百歐維拉提夫治療公司 Factor VIII-XTEN fusions and uses thereof
US10300070B2 (en) 2014-03-27 2019-05-28 The Brigham And Women's Hospital, Inc. Metabolically-activated drug conjugates to overcome resistance in cancer therapy
CN106535826A (en) 2014-06-24 2017-03-22 怡康医疗股份有限公司 Improved metal alloys for medical devices
US9840553B2 (en) 2014-06-28 2017-12-12 Kodiak Sciences Inc. Dual PDGF/VEGF antagonists
CN104434808A (en) 2014-07-03 2015-03-25 石药集团中奇制药技术(石家庄)有限公司 Therapeutic nanoparticles and preparation method thereof
KR101616623B1 (en) * 2014-07-24 2016-04-29 연세대학교 산학협력단 Nanoparticle comprising hydrophobic drug conjugated to cationic polymer and hydrophilic drug conjugated to anionic polymer
CN107405343A (en) 2014-11-07 2017-11-28 明尼苏达大学董事会 Salt and composition available for treatment disease
AR100034A1 (en) 2015-01-30 2016-09-07 Consejo Nac De Investig Científicas Y Técnicas (Conicet) A PHARMACEUTICAL COMPOSITION SOLUBLE IN WATER THAT INCLUDES, AT LEAST, A THERAPEUTICALLY ACTIVE SUBSTANCE AND, AT LEAST, A CAPACITY SUBSTANCE TO FORM MICELAS
WO2017024060A1 (en) 2015-08-03 2017-02-09 Biogen Ma Inc. Factor ix fusion proteins and methods of making and using same
MX2018002226A (en) 2015-08-28 2018-03-23 Amunix Operating Inc Chimeric polypeptide assembly and methods of making and using the same.
JP7088454B2 (en) 2015-12-30 2022-06-21 コディアック サイエンシーズ インコーポレイテッド Antibodies and antibody complexes
WO2017151548A1 (en) 2016-03-04 2017-09-08 Mirus Llc Stent device for spinal fusion
EP3426302B1 (en) 2016-03-10 2022-12-14 IO Therapeutics, Inc. Treatment of autoimmune diseases with combinations of rxr agonists and thyroid hormones
CN109562187B (en) * 2016-03-14 2022-05-17 威斯康星校友研究基金会 Oligolactic acid conjugates and micelles with enhanced anti-cancer efficacy
US10888553B2 (en) * 2016-11-14 2021-01-12 Karlsson-Tuner Invest As Use of non-transition metal coordinated dipyridoxyl compounds to prevent and treat chemotherapy-induced PSN
IT201700067430A1 (en) * 2017-06-16 2018-12-16 Consiglio Nazionale Ricerche Nanoparticles as vehicles for the release of active ingredients and methods for their production
US11517549B2 (en) 2017-09-20 2022-12-06 Io Therapeutics, Inc. Treatment of disease with esters of selective RXR agonists
CA3079121A1 (en) * 2017-10-16 2019-04-25 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Macromolecular platform for targeting scavenger receptor a1
CA3157509A1 (en) 2019-10-10 2021-04-15 Kodiak Sciences Inc. Methods of treating an eye disorder
US20230105243A1 (en) * 2019-12-03 2023-04-06 Onselex Pharmaceuticals, Inc. Polyphosphazene drug carriers
US20240043615A1 (en) * 2020-12-04 2024-02-08 The Methodist Hospital System Amino acid polymer-platinum anticancer drug conjugates
US11896558B2 (en) 2021-12-07 2024-02-13 Io Therapeutics, Inc. Use of an RXR agonist and taxanes in treating Her2+ cancers

Family Cites Families (108)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US41189A (en) * 1864-01-12 Improvement in adjusting logs in saw-mills
US507659A (en) * 1893-10-31 Gottfried knoche
US16285A (en) * 1856-12-23 Manufacture op hosiery
US155992A (en) * 1874-10-13 Improvement in iron pavements
US34363A (en) * 1862-02-11 Improvement in machinery for cleaning cotton
US545880A (en) * 1895-09-10 Cotton chopper and cultivator
US564159A (en) * 1896-07-14 Railway-switch
GB1064417A (en) * 1963-12-09 1967-04-05 Kyowa Hakko Kogyo Kk Process for the preparation of copolypeptides
GB1541435A (en) * 1975-02-04 1979-02-28 Searle & Co Immunological materials
FR2398348A1 (en) * 1977-07-20 1979-02-16 Ferodo Sa POSITIONING DEVICE
US4356166A (en) * 1978-12-08 1982-10-26 University Of Utah Time-release chemical delivery system
US4343579A (en) * 1979-09-07 1982-08-10 Signode Corporation Nail stack
US4485093A (en) * 1982-08-13 1984-11-27 Runge Richard G Immunotoxin conjugate which comprises arsanilic acid, useful for treating malignant tumors, particularly pancreatic cancer
JPS59186924A (en) * 1983-04-08 1984-10-23 Kureha Chem Ind Co Ltd Antitumor agent bonded with human immunoglobulin
DE3463211D1 (en) * 1983-07-01 1987-05-21 Battelle Memorial Institute Biodegradable polypeptide and its use in the sustained release of medicaments
CA1309558C (en) * 1986-05-30 1992-10-27 Quidel D-gl conjugate therapy
US4950713A (en) * 1986-05-30 1990-08-21 La Jolla Pharmaceutical Company Conjugates of D-glutamic acid: D-lysine copolymer and certain biochemicals
IN165717B (en) 1986-08-07 1989-12-23 Battelle Memorial Institute
US5340817A (en) * 1987-04-14 1994-08-23 Research Triangle Institute Method of treating tumors with anti-tumor effective camptothecin compounds
JPS6461422A (en) 1987-09-02 1989-03-08 Nippon Kayaku Kk Water-soluble polymeric carcinostatic agent
JPS6461423A (en) 1987-09-02 1989-03-08 Nippon Kayaku Kk Water-soluble polymeric carcinostatic agent
US4943579A (en) * 1987-10-06 1990-07-24 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Water soluble prodrugs of camptothecin
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
US5169933A (en) 1988-08-15 1992-12-08 Neorx Corporation Covalently-linked complexes and methods for enhanced cytotoxicity and imaging
US4960790A (en) 1989-03-09 1990-10-02 University Of Kansas Derivatives of taxol, pharmaceutical compositions thereof and methods for the preparation thereof
JP2517760B2 (en) * 1989-05-11 1996-07-24 新技術事業団 Water-soluble polymerized pharmaceutical preparation
US5162515A (en) * 1990-01-16 1992-11-10 La Jolla Pharmaceutical Company Conjugates of biologically stable polymers and polynucleotides for treating systemic lupus erythematosus
FR2658076B1 (en) * 1990-02-12 1992-06-12 Sanofi Sa COSMETIC COMPOSITION CONTAINING COPOLYMERS OF AMINO ACIDS, USEFUL AS A MOISTURIZING AGENT.
US5357827A (en) * 1990-03-15 1994-10-25 Abbott Laboratories Torque compensated cam assembly and method
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
JP3310000B2 (en) 1990-11-07 2002-07-29 靖久 桜井 Water-soluble polymer anticancer agent and carrier for drug support
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
JPH05117385A (en) 1991-10-31 1993-05-14 Res Dev Corp Of Japan Production of block copolymer, block copolymer and water-soluble polymeric carcinostatic agent
DE4138042C2 (en) 1991-11-19 1993-10-14 Biotechnolog Forschung Gmbh Epothilones, their production processes and agents containing these compounds
US5272171A (en) 1992-02-13 1993-12-21 Bristol-Myers Squibb Company Phosphonooxy and carbonate derivatives of taxol
PT706373E (en) * 1992-03-23 2000-11-30 Univ Georgetown TAXOL ENCAPSULATED IN A LIPOSOM AND A METHOD
JPH05286868A (en) 1992-04-03 1993-11-02 Kiyoshi Okawa Carcinostatic agent complex and its screening method
US5312922A (en) * 1992-04-06 1994-05-17 Nordion International Inc. Europium and terbium chelators for time-resolved fluorometric assays
AU4406793A (en) 1992-06-04 1993-12-30 Clover Consolidated, Limited Water-soluble polymeric carriers for drug delivery
GB9213077D0 (en) 1992-06-19 1992-08-05 Erba Carlo Spa Polymerbound taxol derivatives
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
US5871710A (en) * 1992-09-04 1999-02-16 The General Hospital Corporation Graft co-polymer adducts of platinum (II) compounds
EP0665729B1 (en) * 1992-09-04 2003-05-07 The General Hospital Corporation Biocompatible polymers containing diagnostic or therapeutic moieties
DE4231408A1 (en) 1992-09-19 1994-03-24 Schleicher & Co Int Device for shredding documents
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
FR2697752B1 (en) * 1992-11-10 1995-04-14 Rhone Poulenc Rorer Sa Antitumor compositions containing taxane derivatives.
US5380751A (en) 1992-12-04 1995-01-10 Bristol-Myers Squibb Company 6,7-modified paclitaxels
MX9308012A (en) 1992-12-24 1994-08-31 Bristol Myers Squibb Co PHOSPHONOOXIMETHYL ETHER OF TAXANE DERIVATIVES, SOLUBLE IN WATER AND PHARMACEUTICAL COMPOSITIONS INCLUDING THEM.
US5981568A (en) 1993-01-28 1999-11-09 Neorx Corporation Therapeutic inhibitor of vascular smooth muscle cells
EP0683676A4 (en) 1993-02-02 1998-09-30 Neorx Corp Directed biodistribution of small molecules.
US5468769A (en) 1993-07-15 1995-11-21 Abbott Laboratories Paclitaxel derivatives
AU693797B2 (en) 1993-07-19 1998-07-09 Angiotech Pharmaceuticals, Inc. Anti-angiogenic compositions and methods of use
US5886026A (en) 1993-07-19 1999-03-23 Angiotech Pharmaceuticals Inc. Anti-angiogenic compositions and methods of use
ES2163453T3 (en) * 1993-09-28 2002-02-01 Hisamitsu Pharmaceutical Co OLIGONUCLEOTIDOS VEHICLE CONSTITUTED BY POLYAMINO ACIDS.
GB9320781D0 (en) * 1993-10-08 1993-12-01 Erba Carlo Spa Polymer-bound camptothecin derivatives
AU8052194A (en) 1993-10-20 1995-05-08 Enzon, Inc. 2'- and/or 7- substituted taxoids
US5880131A (en) 1993-10-20 1999-03-09 Enzon, Inc. High molecular weight polymer-based prodrugs
US5965566A (en) 1993-10-20 1999-10-12 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
US5443953A (en) * 1993-12-08 1995-08-22 Immunomedics, Inc. Preparation and use of immunoconjugates
US5441598A (en) * 1993-12-16 1995-08-15 Motorola, Inc. Polishing pad for chemical-mechanical polishing of a semiconductor substrate
EP0673182B1 (en) * 1994-03-18 2000-03-29 Lg Electronics Inc. Method for automatic control of a microwave oven
US5730968A (en) * 1994-03-31 1998-03-24 Sterling Winthrop Inc. Segmented chelating polymers as imaging and therapeutic agents
US5468754A (en) * 1994-04-19 1995-11-21 Bionumerik Pharmaceuticals, Inc. 11,7 substituted camptothecin derivatives and formulations of 11,7 substituted camptothecin derivatives and methods for uses thereof
US5730990A (en) 1994-06-24 1998-03-24 Enzon, Inc. Non-antigenic amine derived polymers and polymer conjugates
US5629433A (en) * 1994-07-18 1997-05-13 Hauser, Inc. Selective process for the deacylation and deacetylation of taxol and taxanes
US5646159A (en) * 1994-07-20 1997-07-08 Research Triangle Institute Water-soluble esters of camptothecin compounds
US5626862A (en) 1994-08-02 1997-05-06 Massachusetts Institute Of Technology Controlled local delivery of chemotherapeutic agents for treating solid tumors
JP3770628B2 (en) * 1994-08-09 2006-04-26 サントリー株式会社 Prevention and amelioration of medical symptoms through delayed allergic reaction
US5583153A (en) 1994-10-06 1996-12-10 Regents Of The University Of California Use of taxol in the treatment of rheumatoid arthritis
US6295716B1 (en) 1994-10-28 2001-10-02 American Superconductor Corporation Production and processing of (Bi,Pb) SCCO superconductors
US5633573A (en) * 1994-11-10 1997-05-27 Duracell, Inc. Battery pack having a processor controlled battery operating system
US5783178A (en) * 1994-11-18 1998-07-21 Supratek Pharma. Inc. Polymer linked biological agents
US5489589A (en) 1994-12-07 1996-02-06 Bristol-Myers Squibb Company Amino acid derivatives of paclitaxel
US5580899A (en) * 1995-01-09 1996-12-03 The Liposome Company, Inc. Hydrophobic taxane derivatives
US5736156A (en) * 1995-03-22 1998-04-07 The Ohio State University Liposomal anf micellular stabilization of camptothecin drugs
US5866026A (en) * 1995-06-01 1999-02-02 Columbia Machine, Inc. Method and apparatus for accommodating tolerances in a mold for concrete products
US5801191A (en) * 1995-06-01 1998-09-01 Biophysica Foundation Taxoids
AU716005B2 (en) 1995-06-07 2000-02-17 Cook Medical Technologies Llc Implantable medical device
SG50747A1 (en) * 1995-08-02 1998-07-20 Tanabe Seiyaku Co Comptothecin derivatives
US5762909A (en) 1995-08-31 1998-06-09 General Electric Company Tumor targeting with polymeric molecules having extended conformation
US5736129A (en) * 1995-11-17 1998-04-07 Medenica; Rajko D. Flow cytometric pharmacosensitivity assay and method of cancer treatment
CA2192725C (en) * 1995-12-28 2004-04-20 Kenji Tsujihara Camptothecin derivatives
US5776925A (en) 1996-01-25 1998-07-07 Pharmacyclics, Inc. Methods for cancer chemosensitization
US6441025B2 (en) * 1996-03-12 2002-08-27 Pg-Txl Company, L.P. Water soluble paclitaxel derivatives
CZ297979B6 (en) * 1996-03-12 2007-05-16 Pg-Txl Company, L. P. Composition comprising anti-tumor medicament conjugated to water-soluble polymer, its use in the preparation of a medicament and implantable medical device
US5626882A (en) * 1996-03-15 1997-05-06 Agraquest, Inc. Method of using EMU oil as an insect repellent
US6030941A (en) * 1996-05-01 2000-02-29 Avi Biopharma, Inc. Polymer composition for delivering substances in living organisms
US5646506A (en) * 1996-08-21 1997-07-08 Suzuki; Takeshi Method of charging a secondary battery and an apparatus therefor
US5888026A (en) * 1997-01-24 1999-03-30 Her Majesty The Queen In Right Of Canada, As Represented By The Minister Of Natural Resources Backfill paste production facility and method and apparatus for producing high density slurry and paste backfills
US5854006A (en) * 1997-03-28 1998-12-29 The University Of Virginia Gamma-glutamyl transpeptidase-specific antibody, prodrugs for the treatment of gamma-glutamyl transpeptidase-expressing tumors, and methods of administration thereof
US5854382A (en) 1997-08-18 1998-12-29 Meadox Medicals, Inc. Bioresorbable compositions for implantable prostheses
GB9721069D0 (en) 1997-10-03 1997-12-03 Pharmacia & Upjohn Spa Polymeric derivatives of camptothecin
US6011042A (en) * 1997-10-10 2000-01-04 Enzon, Inc. Acyl polymeric derivatives of aromatic hydroxyl-containing compounds
US5960790A (en) * 1997-12-22 1999-10-05 Rich; Albert Clark Modular solar energy collection system
JPH11241027A (en) * 1998-02-26 1999-09-07 Sony Corp Polymeric composite material and its production
US6156017A (en) * 1998-08-11 2000-12-05 Shieh; Shin Jiu Cleaning device
US6218367B1 (en) 1998-09-15 2001-04-17 Organomed Corporation Paclitaxel-carbohydrate conjugates: design, synthesis and biological evaluations
US20010041189A1 (en) 1999-04-13 2001-11-15 Jingya Xu Poly(dipeptide) as a drug carrier
US6191119B1 (en) * 1999-10-15 2001-02-20 Supergen, Inc. Combination therapy including 9-nitro-20(S)-camptothecin
PL358335A1 (en) * 2000-03-17 2004-08-09 Cell Therapeutics, Inc. Polyglutamic acid-camptothecin conjugates and methods of preparation
FR2822834B1 (en) * 2001-04-02 2005-02-25 Flamel Tech Sa COLLOIDAL SUSPENSION OF NANOPARTICLES BASED ON AMPHIPHILIC COPOLYMERS FOR VECTORIZATION OF ACTIVE INGREDIENTS AND THEIR METHOD OF PREPARATION
US6837673B2 (en) * 2002-01-25 2005-01-04 Hitachi, Ltd. Turbine-unit disassembling method and turbine unit

Cited By (227)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8469943B2 (en) 1995-06-07 2013-06-25 Cook Medical Technologies Llc Coated implantable medical device
US7811622B2 (en) 1995-06-07 2010-10-12 Cook Incorporated Coated implantable medical device
US20030176320A1 (en) * 1996-03-12 2003-09-18 Chun Li Water soluble paclitaxel derivatives
US20040018960A1 (en) * 1996-03-12 2004-01-29 Pg-Txl Company, L.P. Water soluble paclitaxel derivatives
US20030114363A1 (en) * 1996-03-12 2003-06-19 Pg-Txl Company, L.P. Water soluble paclitaxel derivatives
US20030130178A1 (en) * 1996-03-12 2003-07-10 Pg-Txl Company, L.P. Water soluble paclitaxel dervatives
US20030130170A1 (en) * 1996-03-12 2003-07-10 Pg-Txl Company, L.P. Water soluble paclitaxel derivatives
US20030147807A1 (en) * 1996-03-12 2003-08-07 Pg-Txl Company, L.P. Water soluble paclitaxel derivatives
US20030114518A1 (en) * 1996-03-12 2003-06-19 Pg-Txl Company, L.P. Water soluble paclitaxel derivatives
US7060724B2 (en) 1996-03-12 2006-06-13 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
US20060287220A1 (en) * 1996-03-12 2006-12-21 Cell Therapeutics, Inc. Water soluble paclitaxel prodrugs
US7384977B2 (en) 1996-03-12 2008-06-10 Pg-Txl Company, L.P. Water soluble paclitaxel prodrugs
US20030113335A1 (en) * 1996-03-12 2003-06-19 Pg-Txl Company, L.P. Water soluble paclitaxel derivatives
US7896912B2 (en) 1998-03-30 2011-03-01 Innovational Holdings, Llc Expandable medical device with S-shaped bridging elements
US7819912B2 (en) 1998-03-30 2010-10-26 Innovational Holdings Llc Expandable medical device with beneficial agent delivery mechanism
US8206435B2 (en) 1998-03-30 2012-06-26 Conor Medsystems, Inc. Expandable medical device for delivery of beneficial agent
US8623068B2 (en) 1998-03-30 2014-01-07 Conor Medsystems, Inc. Expandable medical device with ductile hinges
US8052735B2 (en) 1998-03-30 2011-11-08 Innovational Holdings, Llc Expandable medical device with ductile hinges
US8439968B2 (en) 1998-03-30 2013-05-14 Innovational Holdings, Llc Expandable medical device for delivery of beneficial agent
US8052734B2 (en) 1998-03-30 2011-11-08 Innovational Holdings, Llc Expandable medical device with beneficial agent delivery mechanism
US8361537B2 (en) 1998-03-30 2013-01-29 Innovational Holdings, Llc Expandable medical device with beneficial agent concentration gradient
US7909865B2 (en) 1998-03-30 2011-03-22 Conor Medsystems, LLC Expandable medical device for delivery of beneficial agent
US20070122342A1 (en) * 1999-10-25 2007-05-31 Yang David J 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
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
US20060188438A1 (en) * 1999-10-25 2006-08-24 Yang David J Ethylenedicysteine (EC)-drug conjugates, compositions and methods for tissue specific disease imaging
US20100055035A1 (en) * 1999-10-25 2010-03-04 Yang David J Bisaminoethanethiol-targeting ligand conjugates and compositions
US20050084448A1 (en) * 1999-10-25 2005-04-21 Yang David J. Ethylenedicysteine (EC)-drug conjugates, compositions and methods for tissue specific disease imaging
US8236279B2 (en) 1999-10-25 2012-08-07 Board Of Regents, The University Of Texas System Bisaminoethanethiol-targeting ligand conjugates and compositions
US20050079133A1 (en) * 1999-10-25 2005-04-14 Yang David J. 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
US7615208B2 (en) 1999-10-25 2009-11-10 Board Of Regents, The University Of Texas System Metal ion-labeled bis-aminoethanethiol-targeting ligand 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
US20030211973A1 (en) * 2000-03-17 2003-11-13 Cell Therapeutics, Inc. Polyglutamic acid-camptothecin conjugates and methods of preparation
US20030216289A1 (en) * 2000-03-17 2003-11-20 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
US7153864B2 (en) 2000-03-17 2006-12-26 Cell Therapeutics Inc. Polyglutamic acid-camptothecin conjugates and methods of preparation
US8236048B2 (en) 2000-05-12 2012-08-07 Cordis Corporation Drug/drug delivery systems for the prevention and treatment of vascular disease
US8303609B2 (en) 2000-09-29 2012-11-06 Cordis Corporation Coated medical devices
US6764507B2 (en) 2000-10-16 2004-07-20 Conor Medsystems, Inc. Expandable medical device with improved spatial distribution
US8187321B2 (en) 2000-10-16 2012-05-29 Innovational Holdings, Llc Expandable medical device for delivery of beneficial agent
US7850728B2 (en) 2000-10-16 2010-12-14 Innovational Holdings Llc Expandable medical device for delivery of beneficial agent
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
US9066990B2 (en) 2001-03-26 2015-06-30 Bayer Intellectual Property Gmbh Preparation for restenosis prevention
US8389043B2 (en) 2001-03-26 2013-03-05 Bayer Pharma Aktiengesellschaft Preparation for restenosis prevention
US7750041B2 (en) * 2001-03-26 2010-07-06 Bayer Schering Pharma Aktiengesellschaft Preparation for the prophylaxis of restenosis
US20020197261A1 (en) * 2001-04-26 2002-12-26 Chun Li Therapeutic agent/ligand conjugate compositions, their methods of synthesis and use
US20030003048A1 (en) * 2001-04-26 2003-01-02 Chun Li Diagnostic imaging compositions, their methods of synthesis and use
US8182527B2 (en) 2001-05-07 2012-05-22 Cordis Corporation Heparin barrier coating for controlled drug release
US7850727B2 (en) 2001-08-20 2010-12-14 Innovational Holdings, Llc Expandable medical device for delivery of beneficial agent
US20030232968A1 (en) * 2001-12-21 2003-12-18 Chun Li Dendritic poly (amino acid) carriers and methods of use
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
US9272057B2 (en) 2002-02-26 2016-03-01 Immunomedics, Inc. Combining radioimmunotherapy and antibody-drug conjugates for improved cancer therapy
US8992931B2 (en) 2002-03-01 2015-03-31 Immunomedics, Inc. Combining radioimmunotherapy and antibody-drug conjugates for improved cancer therapy
US9770517B2 (en) 2002-03-01 2017-09-26 Immunomedics, Inc. Anti-Trop-2 antibody-drug conjugates and uses thereof
US20070020337A1 (en) * 2002-03-29 2007-01-25 Maurizio Zenoni Paclitaxel-based antitumor formulation
US20110070156A1 (en) * 2002-06-14 2011-03-24 Immunomedics, Inc. Combining Radioimmunotherapy and Antibody-Drug Conjugates for Improved Cancer Therapy
US8586050B2 (en) 2002-06-14 2013-11-19 Immunomedics, Inc. Combining radioimmunotherapy and antibody-drug conjugates for improved cancer therapy
US8435529B2 (en) 2002-06-14 2013-05-07 Immunomedics, Inc. Combining radioimmunotherapy and antibody-drug conjugates for improved cancer therapy
WO2004011034A1 (en) * 2002-07-31 2004-02-05 General Electric Company Conjugated lysine copolymers for drug delivery and imaging
US9254202B2 (en) 2002-09-20 2016-02-09 Innovational Holdings Llc Method and apparatus for loading a beneficial agent into an expandable medical device
US8257305B2 (en) 2002-09-20 2012-09-04 Bayer Pharma Aktiengesellschaft Medical device for dispensing medicaments
US8349390B2 (en) 2002-09-20 2013-01-08 Conor Medsystems, Inc. Method and apparatus for loading a beneficial agent into an expandable medical device
US8439868B2 (en) 2002-09-20 2013-05-14 Bayer Pharma AG Medical device for dispersing medicaments
US9649476B2 (en) 2002-09-20 2017-05-16 Bayer Intellectual Property Gmbh Medical device for dispersing medicaments
US20040166058A1 (en) * 2002-11-07 2004-08-26 Board Of Regents, The University Of Texas System Ethylenedicysteine (EC)-drug conjugates, compositions and methods for tissue specific disease imaging
US20040143321A1 (en) * 2002-11-08 2004-07-22 Conor Medsystems, Inc. Expandable medical device and method for treating chronic total occlusions with local delivery of an angiogenic factor
US20060002975A1 (en) * 2002-11-08 2006-01-05 Conor Medsystems, Inc. Method and apparatus for reducing tissue damage after ischemic injury
US8425912B2 (en) 2002-12-13 2013-04-23 Immunomedics, Inc. Immunoconjugates with an intracellularly-cleavable linkage
US8080250B1 (en) 2002-12-13 2011-12-20 Immunomedics, Inc. Immunoconjugates with an intracellularly-cleavable linkage
US20060193865A1 (en) * 2002-12-13 2006-08-31 Immunomedics, Inc. Camptothecin-binding moiety conjugates
US8268317B2 (en) 2002-12-13 2012-09-18 Immunomedics, Inc. Camptothecin-binding moiety conjugates
US11077201B2 (en) 2002-12-13 2021-08-03 Immunomedics, Inc. Immunoconjugates with an intracellularly-cleavable linkage
US11066468B2 (en) 2002-12-13 2021-07-20 Immunomedics, Inc. Immunoconjugates with an intracellularly-cleavable linkage
US9233172B2 (en) 2002-12-13 2016-01-12 Immunomedics, Inc. Immunoconjugates with an intracellularly-cleavable linkage
US20100104589A1 (en) * 2002-12-13 2010-04-29 Immunomedics, Inc. Immunoconjugates with an Intracellularly-Cleavable Linkage
US11020489B2 (en) 2002-12-13 2021-06-01 Immunomedics, Inc. Immunoconjugates with an intracellularly-cleavable linkage
US9388237B2 (en) 2002-12-13 2016-07-12 Immunomedics, Inc. Immunoconjugates with an intracellularly-cleavable linkage
US9138485B2 (en) 2002-12-13 2015-09-22 Immunomedics, Inc. Immunoconjugates with an intracellularly-cleavable linkage
US9133268B2 (en) 2002-12-13 2015-09-15 Immunomedics, Inc. Immunoconjugates with an intracellularly-cleavable linkage
US20100015046A1 (en) * 2002-12-13 2010-01-21 Immunomedics, Inc. Camptothecin-Binding Moiety Conjugates
US8420086B2 (en) 2002-12-13 2013-04-16 Immunomedics, Inc. Camptothecin conjugates of anti-CD22 antibodies for treatment of B cell diseases
US10561738B2 (en) 2002-12-13 2020-02-18 Immunomedics, Inc. Immunoconjugates with an intracellularly-cleavable linkage
US8268319B2 (en) 2002-12-13 2012-09-18 Immunomedics, Inc. Immunoconjugates with an intracellularly-cleavable linkage
US9700634B2 (en) 2002-12-13 2017-07-11 Immunomedics, Inc. Immunoconjugates with an intracellularly-cleavable linkage
US20090291093A1 (en) * 2002-12-13 2009-11-26 Immunomedics, Inc. Immunoconjugates with an Intracellularly-Cleavable Linkage
US20040185053A1 (en) * 2002-12-13 2004-09-23 Immunomedics, Inc. Immunoconjugates with an intracellularly-cleavable linkage
USRE48751E1 (en) 2002-12-13 2021-09-28 Immunomedics, Inc. Immunoconjugates with an intracellularly-cleavable linkage
US7999083B2 (en) 2002-12-13 2011-08-16 Immunomedics, Inc. Immunoconjugates with an intracellularly-cleavable linkage
US7585491B2 (en) * 2002-12-13 2009-09-08 Immunomedics, Inc. Immunoconjugates with an intracellularly-cleavable linkage
US10280220B2 (en) 2002-12-13 2019-05-07 Ibc Pharmaceuticals, Inc. Immunoconjugates with an intracellularly-cleavable linkage
US8877901B2 (en) 2002-12-13 2014-11-04 Immunomedics, Inc. Camptothecin-binding moiety conjugates
US8617558B2 (en) 2002-12-13 2013-12-31 Immunomedics, Inc. Camptothecin-binding moiety conjugates
US8834886B2 (en) 2002-12-13 2014-09-16 Immunomedics, Inc. Camptothecin-binding moiety conjugates
US8759496B2 (en) 2002-12-13 2014-06-24 Immunomedics, Inc. Immunoconjugates with an intracellularly-cleavable linkage
US8741300B2 (en) 2002-12-13 2014-06-03 Immunomedics, Inc. Immunoconjugates with an intracellularly-cleavable linkage
US8449901B2 (en) 2003-03-28 2013-05-28 Innovational Holdings, Llc Implantable medical device with beneficial agent concentration gradient
US20070092440A1 (en) * 2003-05-09 2007-04-26 Edgar Braendle Bone localising radiopharmaceutical and tubulin-interacting compound combinatorial
US8197881B2 (en) 2003-09-22 2012-06-12 Conor Medsystems, Inc. Method and apparatus for loading a beneficial agent into an expandable medical device
US20050129619A1 (en) * 2003-12-10 2005-06-16 Board Of Regents, The University Of Texas System N2S2 chelate-targeting ligand conjugates
US9050378B2 (en) 2003-12-10 2015-06-09 Board Of Regents, The University Of Texas System N2S2 chelate-targeting ligand conjugates
US9730891B2 (en) 2004-05-03 2017-08-15 Ipsen Biopharm Ltd. Liposomes useful for drug delivery
US9717723B2 (en) 2004-05-03 2017-08-01 Ipsen Biopharm Ltd. Liposomes useful for drug delivery
US9782349B2 (en) 2004-05-03 2017-10-10 Ipsen Biopharma Ltd. Liposomes useful for drug delivery
US10350201B2 (en) 2004-05-03 2019-07-16 Ipsen Biopharm Ltd. Liposomes useful for drug delivery
US9737528B2 (en) 2004-05-03 2017-08-22 Ipsen Biopharm Ltd. Liposomes useful for drug delivery to the brain
US20160106672A1 (en) * 2004-05-03 2016-04-21 Merrimack Pharmaceuticals, Inc. Liposomes Useful for Drug Delivery
US9724303B2 (en) 2004-05-03 2017-08-08 Ipsen Biopharm Ltd. Liposomes useful for drug delivery
US11052079B2 (en) 2004-05-03 2021-07-06 Ipsen Biopharm Ltd. Liposomes useful for drug delivery
US10722508B2 (en) 2004-05-03 2020-07-28 Ipsen Biopharm Ltd. Liposomes useful for drug delivery
US10413510B2 (en) 2004-05-03 2019-09-17 Ipsen Biopharm Ltd. Liposomes useful for drug delivery
US10988539B2 (en) 2005-03-03 2021-04-27 Immunomedics, Inc. Combination therapy with anti-HLA-DR antibodies and kinase inhibitors in hematopoietic cancers
US20060258736A1 (en) * 2005-05-12 2006-11-16 Bristol-Myers Squibb Company Dosing regimen
US8623371B2 (en) * 2005-07-08 2014-01-07 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Targeting poly-gama-glutamic acid to treat Staphylococcus epidermidis and related infections
US8921071B2 (en) 2005-07-08 2014-12-30 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Targeting poly-gamma-glutamic acid to treat Staphylococcus epidermidis and related infections
US20120301481A1 (en) * 2005-07-08 2012-11-29 National Institutes Of Health Targeting poly-gamma-glutamic acid to treat staphylococcus epidermidis and related infections
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
US20080107598A1 (en) * 2006-10-05 2008-05-08 Yang David J 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
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
US9033919B2 (en) 2006-11-20 2015-05-19 Lutonix, Inc. Medical device rapid drug releasing coatings comprising oils, fatty acids, and/or lipids
US10881644B2 (en) 2006-11-20 2021-01-05 Lutonix, Inc. Treatment of asthma and chronic obstructive pulmonary disease with anti-proliferate and anti-inflammatory drugs
US11534430B2 (en) 2006-11-20 2022-12-27 Lutonix, Inc. Treatment of asthma and chronic obstructive pulmonary disease with anti-proliferate and anti-inflammatory drugs
US9283358B2 (en) 2006-11-20 2016-03-15 Lutonix, Inc. Drug releasing coatings for medical devices
US9289539B2 (en) 2006-11-20 2016-03-22 Lutonix, Inc. Drug releasing coatings for medical devices
US9289537B2 (en) 2006-11-20 2016-03-22 Lutonix, Inc. Medical device rapid drug releasing coatings comprising oils, fatty acids and/or lipids
US9314552B2 (en) 2006-11-20 2016-04-19 Lutonix, Inc. Drug releasing coatings for medical devices
US9314598B2 (en) 2006-11-20 2016-04-19 Lutonix, Inc. Drug releasing coatings for balloon catheters
US8932561B2 (en) 2006-11-20 2015-01-13 Lutonix, Inc. Medical device rapid drug releasing coatings comprising a therapeutic agent and a contrast agent
US10485958B2 (en) 2006-11-20 2019-11-26 Lutonix, Inc. Drug releasing coatings for balloon catheters
US11376404B2 (en) 2006-11-20 2022-07-05 Lutonix, Inc. Drug releasing coatings for medical devices
US9248220B2 (en) 2006-11-20 2016-02-02 Lutonix, Inc. Medical device rapid drug releasing coatings comprising a therapeutic agent and a contrast agent
US10485959B2 (en) 2006-11-20 2019-11-26 Lutonix, Inc. Drug releasing coatings for balloon catheters
US9402935B2 (en) 2006-11-20 2016-08-02 Lutonix, Inc. Treatment of asthma and chronic obstructive pulmonary disease with anti-proliferate and anti-inflammatory drugs
US8425459B2 (en) 2006-11-20 2013-04-23 Lutonix, Inc. Medical device rapid drug releasing coatings comprising a therapeutic agent and a contrast agent
US8366660B2 (en) 2006-11-20 2013-02-05 Lutonix, Inc. Drug releasing coatings for medical devices
US8366662B2 (en) 2006-11-20 2013-02-05 Lutonix, Inc. Drug releasing coatings for medical devices
US9937159B2 (en) 2006-11-20 2018-04-10 Lutonix, Inc. Treatment of asthma and chronic obstructive pulmonary disease with anti-proliferate and anti-inflammatory drugs
US8404300B2 (en) 2006-11-20 2013-03-26 Lutonix, Inc. Drug releasing coatings for medical devices
US8403910B2 (en) 2006-11-20 2013-03-26 Lutonix, Inc. Drug releasing coatings for medical devices
US10994055B2 (en) 2006-11-20 2021-05-04 Lutonix, Inc. Drug releasing coatings for medical devices
US8414909B2 (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
US10912931B2 (en) 2006-11-20 2021-02-09 Lutonix, Inc. Drug releasing coatings for balloon catheters
US10912932B2 (en) 2006-11-20 2021-02-09 Lutonix, Inc. Drug releasing coatings for balloon catheters
US8414525B2 (en) 2006-11-20 2013-04-09 Lutonix, Inc. Drug releasing coatings for medical devices
US8414910B2 (en) 2006-11-20 2013-04-09 Lutonix, Inc. Drug releasing coatings for medical devices
US9694111B2 (en) 2006-11-20 2017-07-04 Lutonix, Inc. Medical device rapid drug releasing coatings comprising a therapeutic agent and a contrast agent
US8998846B2 (en) 2006-11-20 2015-04-07 Lutonix, Inc. Drug releasing coatings for balloon catheters
US9700704B2 (en) 2006-11-20 2017-07-11 Lutonix, Inc. Drug releasing coatings for balloon catheters
US9764065B2 (en) 2006-11-20 2017-09-19 Lutonix, Inc. Drug releasing coatings for medical devices
US10835719B2 (en) 2006-11-20 2020-11-17 Lutonix, Inc. Drug releasing coatings for medical devices
US9023371B2 (en) 2006-11-20 2015-05-05 Lutonix, Inc. Drug releasing coatings for medical devices
US9005161B2 (en) 2006-11-20 2015-04-14 Lutonix, Inc. Drug releasing coatings for medical devices
US9737640B2 (en) 2006-11-20 2017-08-22 Lutonix, Inc. Drug releasing coatings for medical devices
US9737691B2 (en) 2006-11-20 2017-08-22 Lutonix, Inc. Drug releasing coatings for balloon catheters
US8998847B2 (en) 2006-11-20 2015-04-07 Lutonix, Inc. Drug releasing coatings for medical devices
US9757351B2 (en) 2006-11-20 2017-09-12 Lutonix, Inc. Medical device rapid drug releasing coatings comprising oils, fatty acids and/or lipids
US9757544B2 (en) 2006-11-20 2017-09-12 Lutonix, Inc. Drug releasing coatings for medical devices
US9180485B2 (en) 2008-08-29 2015-11-10 Lutonix, Inc. Methods and apparatuses for coating balloon catheters
US9770576B2 (en) 2008-08-29 2017-09-26 Lutonix, Inc. Methods and apparatuses for coating balloon catheters
US8430055B2 (en) 2008-08-29 2013-04-30 Lutonix, Inc. Methods and apparatuses for coating balloon catheters
US9480756B2 (en) 2009-02-13 2016-11-01 Immunomedics, Inc. Immunoconjugates with an intracellularly-cleavable linkage
US9345783B2 (en) 2009-02-13 2016-05-24 Immunomedics, Inc. Immunoconjugates with an intracellularly-cleavable linkage
US9198978B2 (en) 2009-02-13 2015-12-01 Immunomedics, Inc. Immunoconjugates with an intracellularly-cleavable linkage
US10322193B1 (en) 2009-02-13 2019-06-18 Immunomedics, Inc. Immunoconjugates with an intracellularly-cleavable linkage
US9102735B2 (en) 2009-02-13 2015-08-11 Immunomedics, Inc. Immunoconjugates with an intracellularly-cleavable linkage
US10265414B2 (en) 2009-02-13 2019-04-23 Immunomedics, Inc. Immunoconjugates with an intracellularly-cleavable linkage
US9675706B2 (en) 2009-02-13 2017-06-13 Immunomedics, Inc. Immunoconjugates with an intracellularly-cleavable linkage
US8999344B2 (en) 2009-02-13 2015-04-07 Immunomedics, Inc. Immunoconjugates with an intracellularly-cleavable linkage
US10064957B2 (en) 2009-02-13 2018-09-04 Immunomedics, Inc. Immunoconjugates with an intracellularly-cleavable linkage
US20130231292A1 (en) * 2012-03-02 2013-09-05 Wei Zhou Synergistic anti-proliferation activity of TAS-108 with mTOR inhibitors against cancer cells
CN102558270A (en) * 2012-03-09 2012-07-11 辽宁新中现代医药有限公司 20(S) and 20(R)-dammarane-3beta,12beta,20,25-tetrol derivative and preparation method and application thereof
US9382329B2 (en) 2012-08-14 2016-07-05 Ibc Pharmaceuticals, Inc. Disease therapy by inducing immune response to Trop-2 expressing cells
US10662252B2 (en) 2012-08-14 2020-05-26 Ibc Pharmaceuticals, Inc. Disease therapy by inducing immune response to Trop-2 expressing cells
US9879088B2 (en) 2012-08-14 2018-01-30 Ibc Pharmaceuticals, Inc. Disease therapy by inducing immune response to Trop-2 expressing cells
US9670286B2 (en) 2012-08-14 2017-06-06 Ibc Pharmaceuticals, Inc. Disease therapy by inducing immune response to Trop-2 expressing cells
US10130626B2 (en) 2012-12-13 2018-11-20 Immunomedics, Inc. Dosages of immunoconjugates of antibodies and SN-38 for improved efficacy and decreased toxicity
US9493573B2 (en) 2012-12-13 2016-11-15 Immunomedics, Inc. Dosages of immunoconjugates of antibodies and SN-38 for improved efficacy and decreased toxicity
US10206918B2 (en) 2012-12-13 2019-02-19 Immunomedics, Inc. Efficacy of anti-HLA-DR antiboddy drug conjugate IMMU-140 (hL243-CL2A-SN-38) in HLA-DR positive cancers
US11541047B2 (en) 2012-12-13 2023-01-03 Immunomedics, Inc. Dosages of immunoconjugates of antibodies and SN-38 for improved efficacy and decreased toxicity
US10143756B2 (en) 2012-12-13 2018-12-04 Immunomedics, Inc. Antibody-SN-38 immunoconjugates with a CL2A linker
US10413539B2 (en) 2012-12-13 2019-09-17 Immunomedics, Inc. Therapy for metastatic urothelial cancer with the antibody-drug conjugate, sacituzumab govitecan (IMMU-132)
US10137196B2 (en) 2012-12-13 2018-11-27 Immunomedics, Inc. Dosages of immunoconjugates of antibodies and SN-38 for improved efficacy and decreased toxicity
US9226973B2 (en) 2012-12-13 2016-01-05 Immunomedics, Inc. Dosages of immunoconjugates of antibodies and SN-38 for improved efficacy and decreased toxicity
US9375489B2 (en) 2012-12-13 2016-06-28 Immunomedics, Inc. Antibody-SN-38 immunoconjugates with a CL2A linker
US10130718B2 (en) 2012-12-13 2018-11-20 Immunomedics, Inc. Antibody-drug conjugates and uses thereof
US9458242B2 (en) 2012-12-13 2016-10-04 Immunomedics, Inc. Dosages of immunoconjugates of antibodies and SN-38 for improved efficacy and decreased toxicity
US10034950B2 (en) 2012-12-13 2018-07-31 Immunomedics, Inc. Antibody-SN-38 immunoconjugates with a CL2A linker
US10653793B2 (en) 2012-12-13 2020-05-19 Immunomedics, Inc. Antibody-drug conjugates and uses thereof
US9931417B2 (en) 2012-12-13 2018-04-03 Immunomedics, Inc. Antibody-SN-38 immunoconjugates with a CL2A linker
US10682347B2 (en) 2012-12-13 2020-06-16 Immunomedics, Inc. Dosages of immunoconjugates of antibodies and SN-38 for improved efficacy and decreased toxicity
US10709701B2 (en) 2012-12-13 2020-07-14 Immunomedics, Inc. Efficacy of anti-HLA-DR antibody drug conjugate IMMU-140 (hL243-CL2A-SN-38) in HLA-DR positive cancers
US9855344B2 (en) 2012-12-13 2018-01-02 Immunomedics, Inc. Antibody-SN-38 immunoconjugates with a CL2A linker
US10744129B2 (en) 2012-12-13 2020-08-18 Immunomedics, Inc. Therapy of small-cell lung cancer (SCLC) with a topoisomerase-I inhibiting antibody-drug conjugate (ADC) targeting Trop-2
US10751420B2 (en) 2012-12-13 2020-08-25 Immunomedics, Inc. Antibody-SN-38 immunoconjugates with a CL2A linker
US11116846B2 (en) 2012-12-13 2021-09-14 Immunomedics, Inc. Antibody-drug conjugates and uses thereof
US9475884B2 (en) 2012-12-13 2016-10-25 Immunomedics, Inc. Dosages of immunoconjugates of antibodies and SN-38 for improved efficacy and decreased toxicity
US9028833B2 (en) 2012-12-13 2015-05-12 Immunomedics, Inc. Dosages of immunoconjugates of antibodies and SN-38 for improved efficacy and decreased toxicity
US10849986B2 (en) 2012-12-13 2020-12-01 Immunomedics, Inc. Antibody-drug conjugates and uses thereof
US9481732B2 (en) 2012-12-13 2016-11-01 Immunomedics, Inc. Dosages of immunoconjugates of antibodies and SN-38 for improved efficacy and decreased toxicity
US9107960B2 (en) 2012-12-13 2015-08-18 Immunimedics, Inc. Antibody-SN-38 immunoconjugates with a CL2A linker
US9629926B2 (en) 2012-12-13 2017-04-25 Immunomedics, Inc. Antibody-SN-38 immunoconjugates with a CL2A linker
US10918721B2 (en) 2012-12-13 2021-02-16 Immunomedics, Inc. Dosages of immunoconjugates of antibodies and SN-38 for improved efficacy and decreased toxicity
US11052156B2 (en) 2012-12-13 2021-07-06 Immunomedics, Inc. Antibody-drug conjugates and uses thereof
US9522959B2 (en) 2012-12-13 2016-12-20 Immunomedics, Inc. Dosages of immunoconjugates of antibodies and SN-38 for improved efficacy and decreased toxicity
US11052081B2 (en) 2012-12-13 2021-07-06 Immunomedics, Inc. Therapy for metastatic urothelial cancer with the antibody-drug conjugate, sacituzumab govitecan (IMMU-132)
US9499631B2 (en) 2012-12-13 2016-11-22 Immunomedics, Inc. Dosages of immunoconjugates of antibodies and SN-38 for improved efficacy and decreased toxicity
US9493574B2 (en) 2012-12-13 2016-11-15 Immunomedics, Inc. Dosages of immunoconjugates of antibodies and SN-38 for improved efficacy and decreased toxicity
US9492566B2 (en) 2012-12-13 2016-11-15 Immunomedics, Inc. Antibody-drug conjugates and uses thereof
US9707302B2 (en) 2013-07-23 2017-07-18 Immunomedics, Inc. Combining anti-HLA-DR or anti-Trop-2 antibodies with microtubule inhibitors, PARP inhibitors, bruton kinase inhibitors or phosphoinositide 3-kinase inhibitors significantly improves therapeutic outcome in cancer
US11253606B2 (en) 2013-07-23 2022-02-22 Immunomedics, Inc. Combining anti-HLA-DR or anti-Trop-2 antibodies with microtubule inhibitors, PARP inhibitors, Bruton kinase inhibitors or phosphoinositide 3-kinase inhibitors significantly improves therapeutic outcome in cancer
US10436788B2 (en) 2015-04-22 2019-10-08 Immunomedics, Inc. Isolation, detection, diagnosis and/or characterization of circulating Trop-2-positive cancer cells
US9797907B2 (en) 2015-04-22 2017-10-24 Immunomedics, Inc. Isolation, detection, diagnosis and/or characterization of circulating Trop-2-positive cancer cells
US10195175B2 (en) 2015-06-25 2019-02-05 Immunomedics, Inc. Synergistic effect of anti-Trop-2 antibody-drug conjugate in combination therapy for triple-negative breast cancer when used with microtubule inhibitors or PARP inhibitors
US10058621B2 (en) 2015-06-25 2018-08-28 Immunomedics, Inc. Combination therapy with anti-HLA-DR antibodies and kinase inhibitors in hematopoietic cancers
US11439620B2 (en) 2015-06-25 2022-09-13 Immunomedics, Inc. Synergistic effect of anti-trop-2 antibody-drug conjugate in combination therapy for triple-negative breast cancer when used with microtubule inhibitors or PARP inhibitors
US10993914B2 (en) 2015-10-16 2021-05-04 Ipsen Biopharm Ltd. Stabilizing camptothecin pharmaceutical compositions
US10456360B2 (en) 2015-10-16 2019-10-29 Ipsen Biopharm Ltd. Stabilizing camptothecin pharmaceutical compositions
US10954305B2 (en) 2016-02-10 2021-03-23 Immunomedics, Inc. Combination of ABCG2 inhibitors with sacituzumab govitecan (IMMU-132) overcomes resistance to SN-38 in Trop-2 expressing cancers
US10266605B2 (en) 2016-04-27 2019-04-23 Immunomedics, Inc. Efficacy of anti-trop-2-SN-38 antibody drug conjugates for therapy of tumors relapsed/refractory to checkpoint inhibitors
US11192955B2 (en) 2016-04-27 2021-12-07 Immunomedics, Inc. Efficacy of anti-Trop-2-SN-38 antibody drug conjugates for therapy of tumors relapsed/refractory to checkpoint inhibitors
US10918734B2 (en) 2017-03-27 2021-02-16 Immunomedics, Inc. Treatment of high Trop-2 expressing triple negative breast cancer (TNBC) with sacituzumab govitecan (IMMU-132) overcomes homologous recombination repair (HRR) rescue mediated by Rad51
US10799597B2 (en) 2017-04-03 2020-10-13 Immunomedics, Inc. Subcutaneous administration of antibody-drug conjugates for cancer therapy

Also Published As

Publication number Publication date
CA2303338A1 (en) 1999-10-07
US20030124055A1 (en) 2003-07-03
US20060287220A1 (en) 2006-12-21
US7135496B2 (en) 2006-11-14
DE69927350D1 (en) 2006-02-02
US20030114518A1 (en) 2003-06-19
US20060135404A1 (en) 2006-06-22
US20030166507A1 (en) 2003-09-04
US20030114363A1 (en) 2003-06-19
ATE304867T1 (en) 2005-10-15
US20030130341A1 (en) 2003-07-10
DE69927350T2 (en) 2006-06-22
US6730699B2 (en) 2004-05-04
DK1028756T3 (en) 2006-02-20
EP1598081A3 (en) 2008-05-07
US20030147807A1 (en) 2003-08-07
US20030114397A1 (en) 2003-06-19
US20030073615A1 (en) 2003-04-17
US7060724B2 (en) 2006-06-13
AU3455699A (en) 1999-10-18
US20030130170A1 (en) 2003-07-10
US20030073617A1 (en) 2003-04-17
US20040018960A1 (en) 2004-01-29
US20060111273A1 (en) 2006-05-25
ES2249888T3 (en) 2006-04-01
US20040198638A1 (en) 2004-10-07
US6515017B1 (en) 2003-02-04
EP1028756A1 (en) 2000-08-23
US7384977B2 (en) 2008-06-10
US20030134793A1 (en) 2003-07-17
CA2303338C (en) 2011-01-04
EP1028756B1 (en) 2005-09-21
US6441025B2 (en) 2002-08-27
US20080153865A1 (en) 2008-06-26
TWI246922B (en) 2006-01-11
EP1028756A4 (en) 2002-05-08
WO1999049901A1 (en) 1999-10-07
TWI255183B (en) 2006-05-21
MY121807A (en) 2006-02-28
US20030130178A1 (en) 2003-07-10
US20030176320A1 (en) 2003-09-18
US20030113335A1 (en) 2003-06-19
EP1598081A2 (en) 2005-11-23
TW200517105A (en) 2005-06-01
US6884817B2 (en) 2005-04-26

Similar Documents

Publication Publication Date Title
US6441025B2 (en) Water soluble paclitaxel derivatives
AU735900B2 (en) Water soluble paclitaxel prodrugs
JP2001288097A (en) Water-soluble paclitaxel derivative
Copie et al. Water soluble paclitaxel prodrugs
MXPA98007442A (en) Soluble paclitaxel profarmacos in a

Legal Events

Date Code Title Description
AS Assignment

Owner name: PG-TXL COMPANY, L.P., TEXAS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:LI, CHUN;WALLACE, SIDNEY;YU, DONG-FANG;AND OTHERS;REEL/FRAME:009847/0336

Effective date: 19990201

STCF Information on status: patent grant

Free format text: PATENTED CASE

FEPP Fee payment procedure

Free format text: PAYOR NUMBER ASSIGNED (ORIGINAL EVENT CODE: ASPN); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

FPAY Fee payment

Year of fee payment: 4

FEPP Fee payment procedure

Free format text: PAT HOLDER NO LONGER CLAIMS SMALL ENTITY STATUS, ENTITY STATUS SET TO UNDISCOUNTED (ORIGINAL EVENT CODE: STOL); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

REFU Refund

Free format text: REFUND - PAYMENT OF MAINTENANCE FEE, 8TH YR, SMALL ENTITY (ORIGINAL EVENT CODE: R2552); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

FPAY Fee payment

Year of fee payment: 8

FPAY Fee payment

Year of fee payment: 12