USRE41393E1 - Treatment of refractory tumors using epothilone derivatives - Google Patents

Treatment of refractory tumors using epothilone derivatives Download PDF

Info

Publication number
USRE41393E1
USRE41393E1 US11/346,579 US34657906A USRE41393E US RE41393 E1 USRE41393 E1 US RE41393E1 US 34657906 A US34657906 A US 34657906A US RE41393 E USRE41393 E US RE41393E
Authority
US
United States
Prior art keywords
methyl
dihydroxy
thiazolyl
ethenyl
dione
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Expired - Lifetime
Application number
US11/346,579
Inventor
Francis Y.F. Lee
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.)
R-PHARM US OPERATING LLC
Original Assignee
Bristol Myers Squibb Co
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
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=23028846&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=USRE41393(E1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Bristol Myers Squibb Co filed Critical Bristol Myers Squibb Co
Priority to US11/346,579 priority Critical patent/USRE41393E1/en
Application granted granted Critical
Publication of USRE41393E1 publication Critical patent/USRE41393E1/en
Assigned to R-PHARM US OPERATING LLC reassignment R-PHARM US OPERATING LLC ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: BRISTOL-MYERS SQUIBB COMPANY
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
    • 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
    • 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
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/425Thiazoles
    • A61K31/427Thiazoles not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents

Definitions

  • the present invention relates to the use of certain potent epothilone analogs in the treatment of tumors that have demonstrated resistance to therapy with other chemotherapeutic agents.
  • Epothiolones are macrolide compounds that find utility in the pharmaceutical field.
  • epothilones A and B having the structures: may be found to exert microtubule-stabilizing effects similar to paclitaxel (TAXOL®) and hence cytotoxic activity against rapidly proliferating cells, such as tumor cells or other hyperproliferative cellular diseases.
  • TAXOL® paclitaxel
  • cytotoxic activity against rapidly proliferating cells such as tumor cells or other hyperproliferative cellular diseases.
  • epothilones A and B have been synthesized and may be used to treat a variety of cancers and other abnormal proliferative diseases. Such analogs are disclosed in Hofle et al., Id.; Nicolaou et al., Agnew. Chem. Int. Ed. Eng., Vol.36, No. 19, 2097-2103 (1997); and Su et al., Agnew. Chem. Int. Ed. Engl., Vol. 36, No. 19, 2093-2097 (1997). In some instances, epothilone derivatives have demonstrated enhanced properties over epothilones A and B. The present invention is concerned with the discovery that certain epothilone derivatives may be utilized to treat cancers that have demonstrated resistance to other chemotherapeutic agents, such as oncolytic agents of the taxane family of compounds.
  • chemotherapeutic agents such as oncolytic agents of the taxane family of compounds.
  • tumors demonstrating a clinical resistance to treatment with other chemotherapeutic agents may be treated with an epothilone derivative selected from those represented by formula I: wherein B 1 , B 2 , G, Q, X, Y, Z 1 , Z 2 , and R 1 through R 7 have the meanings given below.
  • the compounds represented by formula I have previously demonstrated significantly enhanced potency over known chemotherapeutic agents, for example, epothilones A and B above and certain others including those in the taxane series.
  • Compounds represented by formula I are further advantageous in that, unlike most oncology agents, they are efficacious via oral administration.
  • FIG. 1 is a bar graph showing the cytotoxicity spectrum of a compound of the invention against a panel of tumor cell lines.
  • FIG. 2 is a bar graph showing the cytotoxicity of a compound of the invention against paclitaxel-resistant tumors.
  • FIG. 3 shows the mitotic blockade induced by a compound of the invention.
  • Processes of the present invention provide advantageous treatment for tumors that have demonstrated resistance to treatment with chemotherapeutic agents, such as those of the taxane family.
  • the term “resistance to treatment” as utilized herein includes both tumors that are initially unresponsive to treatment with a chemotherapeutic agent as well as tumors that are initially responsive, but develop resistance over the course of treatment.
  • Compounds useful in the subject method are epothilones, a class of oncology chemotherapeutic agents chemically distinct from the taxane family of oncology agents.
  • the subject epothilone derivatives are represented by formula I: wherein
  • Preferred compounds in accordance with the present invention are those represented by formula I above wherein Q is
  • a particular preferred compound in accordance with the present invention is represented by the formula: This compound chemically is [1S-[1R*,3R*(E),7R*,10S*,11R*,12R*,16S*]]-7,11-dihydroxy-8,8,10,12,16-pentamethyl-3-[1-methyl-2-(2-methyl-4-thiazolyl)ethenyl]-4-aza-17-oxabicyclo[14.1.0]heptadecane-5,9-dione.
  • alkyl refers to optionally substituted straight- or branched-chain saturated hydrocarbon groups having from 1 to about 20 carbon atoms, preferably from 1 to about 7 carbon atoms.
  • lower alkyl refers to optionally substituted alkyl groups having from 1 to about 4 carbon atoms.
  • substituted alkyl refers to an alkyl group substituted by, for example, one to four substituents, such as, halo, trifluoromethyl, trifluoromethoxy, hydroxy, alkoxy, cycloalkyoxy, heterocylooxy, oxo, alkanoyl, aryl, aryloxy, aralkyl, alkanoyloxy, amino, alkylamino, arylamino, aralkylamino, cycloalkylamino, heterocycloamino, disubstituted amino in which the two substituents on the amino group are selected from alkyl, aryl, aralkyl, alkanoylamino, aroylamino, aralkanoylamino, substituted alkanoylamino, substituted arylamino, substituted aralkanoylamino, thiol, alkylthio, arylthio, a
  • substituents themselves are further substituted, such further substituents are selected from the group consisting of halogen, alkyl, alkoxy, aryl and aralkyl.
  • alkyl and substituted alkyl apply as well to the alkyl portion of alkoxy groups.
  • alkenyl refers to optionally substituted unsaturated aliphatic hydrocarbon groups having from 1 to about 9 carbons and one or more double bonds. Substituents may include one or more substituent groups as described above for substituted alkyl.
  • halogen refers to fluorine, chlorine, bromine and iodine.
  • ring system refers to an optionally substituted ring system containing one to three rings and at least one carbon to carbon double bond in at least one ring.
  • exemplary ring systems include, but are not limited to, an aryl or a partially or fully unsaturated heterocyclic ring system, which may be optionally substituted.
  • aryl refers to monocyclic or bicyclic aromatic hydrocarbon groups having from about 6 to about 12 carbon atoms in the ring portion, for example, phenyl, naphthyl, biphenyl and diphenyl groups, each of which may be substituted.
  • aralkyl refers to an aryl group bonded to a larger entity through an alkyl group, such as benzyl.
  • substituted aryl refers to an aryl group substituted by, for example, one to four substituents such as alkyl; substituted alkyl, halo, trifluoromethyl, trifluoromethoxy, hydroxy, alkoxy, cycloalkyloxy, heterocyclooxy, alkanoyl, alkanyloxy, amino, alkylamino, dialkylamino, aralkylamino, cycloalkylamino, heterocycloamino, alkanoylamino, thiol, alkylthio, cycloalkylthio, heterocyclothio, ureido, nitro, cyano, carboxy, carboxyalkyl, carbamyl, alkoxycarbonyl, alkylthio, arylthiono, alkylsulfonyl, sulfonamido, aryloxy and the like.
  • the substituent may be further substituted by one or more members selected from
  • cycloalkyl refers to optionally substituted saturated cyclic hydrocarbon ring systems, preferably containing 1 to about 3 rings and 3 to about 7 carbon atoms per ring, which may be further fused with an unsaturated C 3 -C 7 carbocyclic ring.
  • exemplary groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, cyclodecyl, cyclododecyl, and adamantyl.
  • substituents include one or more alkyl groups as described above, or one or more of the groups described above as substituents for alkyl groups.
  • heterocycle refers to an optionally substituted, unsaturated, partially saturated, or fully saturated, aromatic or nanoaromatic cyclic group, for example, which is a 4- to 7-membered monocyclic, 7-to 11-membered bicyclic, or 10- to 15-membered tricyclic ring system, which has at least one heteroatom in at least one carbon atom-containing ring.
  • Each ring of the heterocyclic group containing a heteroatom may have 1, 2 or 3 heteroatoms selected from nitrogen atoms, oxygen atoms and sulfur atoms, where the nitrogen and sulfur heteroatoms may also optionally be oxidized and the nitrogen heteroatoms may also optionally be quaternized.
  • the heterocyclic group may be attached at any heteroatom or carbon atom.
  • Exemplary monocyclic heterocyclic groups include pyrrolidinyl, pyrrolyl, indolyl, pyrazolyl, oxetanyl, pyrazolinyl, imidazolyl, imidazolinyl, imidazolidinyl, oxazolyl, oxazolidinyl, isoxazolinyl, isoxazolyl, thiazolyl, thiadiazolyl, thiazolidinyl, isothiazolyl, isothiazolidinyl, furyl, tetrahydrofuryl, thienyl, oxadiazolyl, piperidinyl, piperizinyl, 2-oxopiperazinyl, 2-oxopiperidinyl, 2-oxopyrrolidinyl, 2-oxazepinyl, azepinyl, 4-piperidonyl, pyridyl, N-oxo-pyridyl,
  • bicyclic heterocyclic groups include benzothiazolyl, benzoxazolyl, benzothienyl, quinuclidinyl, quinolinyl, quinolinyl-N-oxide, tetrahydroisoquinolinyl, isoquinolinyl, benzimidazolyl, benzopyranyl, indolizinyl, benzofuryl, chromonyl, coumarinyl, cinnolinyl, quinoxalinyl, indazolyl, pyrrolopyridyl, furopyridinyl (such as furo[2,3-c]pyridinyl, furo[3,1-b]pyridinyl] or furo[2,3-b]pyridinyl), dihydroisoindolyl, dihydroquinazolinyl (such as 3,4-dihydro-4-oxo-quinazolinyl), benzisothiazolyl,
  • substituents for the terms “ring system,” “heterocycle,” “heterocyclic,” and “heterocyclo” include one or more substituent groups as described above for substituted alkyl or substituted aryl, and smaller heterocycles, such as, epoxides, aziridines and the like.
  • alkanoyl refers to —C(O)-alkyl.
  • substituted alkanoyl refers to —C(O)-substituted alkyl.
  • heteroatoms shall include oxygen, sulfur and nitrogen.
  • the compounds represented by formula I form salts with a variety of organic and inorganic acids.
  • Such salts include those formed with hydrogen chloride, hydrogen bromide, methanesulfonic acid, hydroxyethanesulfonic acid, sulfuric acid, acetic acid, trifluoroacetic acid, maleic acid, benzenesulfonic acid, toluenesulfonic acid and various others as are recognized by those of ordinary skill in the art of pharmaceutical compounding.
  • Such salts are formed by reacting a compound represented by formula I in an equivalent amount of the acid in a medium in which the salt precipitates or in an aqueous medium followed by evaporation.
  • zwitterions inner salts
  • solvates and hydrates of the compounds represented by formula I are also included herein.
  • the compounds represented by formula I above may exist as multiple optical, geometric, and stereoisomers. While the compounds shown herein are depicted for one optical orientation, included within the present invention are all isomers and mixtures thereof.
  • the compounds represented by formula I above are microtubule-stabilizing agents. Therefore, they are useful in the treatment of a variety of cancers and other proliferative diseases including, but not limited to, the following:
  • Oncology therapy refers to treatment of cancer or tumors with chemotherapeutic agents that exert a cytotoxic effect in cells.
  • An example of a chemotherapeutic agent is an oncology agent of the taxane family of compounds. It is known, for example, that a considerable number of patients initially responsive to oncology therapy with taxane compounds develop resistance over a course of therapy and that not all cancers respond to treatment with taxane therapy as is the case with virtually all oncology agents. Further, certain diseases, such as cholorectal cancers or melanoma, are known to be innately resistant to taxane therapy.
  • the subject epothilone compounds are highly potent cytotoxic agents capable of killing cancer cells at low nanometer concentrations and are approximately twice as potent as paclitaxel in inducing tubulin polymerization. More important, the subject compound seem to possess the capacity to retain their antineoplastic activity against human cancers that are naturally insensitive to paclitaxel or have developed resistance to it, both in vitro and in vivo.
  • Tumors for which the subject epothilone compounds have demonstrated significant antitumor activity include, without intended limitation, the following:
  • the compounds represented by formula I have demonstrated that they are orally efficacious versus preclinical human tumor xenografts grown in immunocompromized mice or rats. Being efficacious upon oral administration is considered a significant advantage of the subject epothilone derivatives.
  • the present invention thus provides a method of treating a subject, preferably mammals and especially humans, in need of treatment for a tumor that has demonstrated resistance to therapy with the taxane family of oncologic agents, comprising administering to the subject one of the epothilone compounds represented by formula I in an amount effective for such treatment.
  • Other therapeutic agents such as those described below, may be employed with the subject epothilone compounds in their usual dosages. Such agents may be administered prior to, simultaneously with, or following the subject epothilone compounds.
  • An effective amount of the epothilone compounds represented by formula I may be determined by one of ordinary skill in the art, and includes exemplary dosage amounts for a human of from about 0.05 to about 200 mg/kg/day. This dosage is typically administered in a single dose, but can be administered in divided doses since the subject compounds are advantageously efficacious via oral administration.
  • the compounds may be administered in a frequent regimen, e.g., every two days for five doses, or intermittently, e.g., every four days for three doses or every eight days for three doses.
  • compositions containing an amount thereof effective for cancer therapy, and a pharmaceutically acceptable carrier.
  • Such compositions may contain other therapeutic agents as described below, and may be formulated, for example, by employing conventional solid or liquid vehicles or diluents, as well as pharmaceutical additives of a type appropriate to the mode of desired administration (for example, excipients, binders, preservatives, stabilizers, flavors, etc.) according to techniques such as those well known in the art of pharmaceutical formulation and/or called for accepted pharmaceutical practice.
  • the compounds represented by formula I may be administered by any suitable means, for example, orally, such as in the form of tablets, capsules, granules or powders; sublingually; bucally; parenterally, such as by subcutaneous, intravenous, intramuscular, or intrasternal injection or infusion techniques (e.g., as sterile injectable aqueous or non-aqueous solutions or suspensions); nasally, such as by inhalation spray; topically, such as in the form of a cream or ointment; or rectally such as in the form of suppositories; in dosage unit formulations containing non-toxic, pharmaceutically acceptable vehicles or diluents.
  • suitable means for example, orally, such as in the form of tablets, capsules, granules or powders; sublingually; bucally; parenterally, such as by subcutaneous, intravenous, intramuscular, or intrasternal injection or infusion techniques (e.g., as sterile injectable aqueous or non-
  • the subject compounds may, for example, be administered in a form suitable for immediate release or extended release. Immediate release or extended release may be achieved by the use of suitable pharmaceutical compositions comprising the present compounds, or, particularly in the case of extended release, by the use of devices such as subcutaneous implants or osmotic pumps.
  • the subject compounds may also be administered liposomally.
  • Suitable dosage forms for the subject epothilone derivatives include, without intended limitation, a orally effective composition such as a tablet, capsule, solution or suspension containing about 5 to about 500 mg per unit dosage of a compound represented by formula I or a topical form (about 0.01% to about 5% by weight compound represented by formula I, one to five treatments per day). They may be compounded in a conventional manner with a physiologically acceptable vehicle or carrier, excipient, binder, preservative, stabilizer, flavor, etc., or with a topical carrier. The compounds represented by formula I can also be formulated in compositions such as sterile solutions or suspensions for parenteral administration.
  • a compound represented by formula I may be compounded with a physiologically acceptable vehicle, carrier, excipient, binder preservative, stabilizer, etc., in a unit dosage form as called for by accepted pharmaceutical practice.
  • the amount of active substance in these compositions or preparations is preferably such that a suitable dosage in the range indicated is obtained.
  • compositions for oral administration include suspensions which may contain, for example, microcrystalline cellulose for imparting bulk, alginic acid or sodium alginate as a suspending agent, methylcellulose as a viscosity enhancer, and sweeteners, or flavoring agents such as those known in the art; and immediate release tablets which may contain, for example, microcrystalline cellulose, dicalcium phosphate, starch, magnesium stearate and/or lactose and/or other excipients, binders, extenders, disintegrants, diluents and lubricants such as those known in the art. Molded tablets, compressed tablets or freeze-dried tablets are exemplary forms that may be used.
  • compositions include those formulating the present compound(s) with fast dissolving diluents such as mannitol, lactose, sucrose and/or cyclodextrins. Also included in such formulations may be high molecular weight excipients such as celluloses (Avicel) or polyethylene glycols (PEG). Such formulations may also include an excipient to aid mucosal adhesion such as hydroxy propyl cellulose (HPC), hydroxy propyl methyl cellulose (HPMC), sodium carboxy methyl cellulose (SCMC), maleic anhydride copolymer (e.g. Gantrez), and agents to control release such as polyacrylic acid copolymer (e.g. Carbopol 934). Lubricants, glidants, flavors, coloring agents and stabilizers may also be added for ease of fabrication and use.
  • fast dissolving diluents such as mannitol, lactose, sucrose and/or cyclodextrins
  • compositions for nasal aerosol or inhalation administration include solutions in saline which may contain, for example, benzyl alcohol or other suitable preservatives, absorption promoters to enhance bioavailability, and/or other solubilizing or dispersing agents such as those known in the art.
  • compositions for parenteral administration include injectable solutions or suspensions which may contain, for example, suitable non-toxic, parentally acceptable diluents or solvents, such as Cremophor® (polyoxyethylated caster oil surfactant), mannitol, 1,3-butanediol, water, Ringer's solution, Lactated Ringer's solution, an isotonic sodium chloride solution, or other suitable dispersion or wetting and suspending agents, including synthetic mono- or diglycerides, and fatty acids, including oleic acid.
  • suitable non-toxic, parentally acceptable diluents or solvents such as Cremophor® (polyoxyethylated caster oil surfactant), mannitol, 1,3-butanediol, water, Ringer's solution, Lactated Ringer's solution, an isotonic sodium chloride solution, or other suitable dispersion or wetting and suspending agents, including synthetic mono- or diglycerides, and
  • compositions for rectal administration include suppositories which may contain, for example, a suitable non-irritating excipient, such as cocoa butter, synthetic glyceride esters or polyethylene glycols, which are solid at ordinary temperature, but liquefy and/or dissolve in the rectal cavity to release the drug.
  • a suitable non-irritating excipient such as cocoa butter, synthetic glyceride esters or polyethylene glycols, which are solid at ordinary temperature, but liquefy and/or dissolve in the rectal cavity to release the drug.
  • the compounds of the invention may be administered either alone or in combination with other chemotherapeutic agents or anti-cancer and cytotoxic agents and/or treatments useful in the treatment of cancer or other proliferative diseases.
  • chemotherapeutic agents or anti-cancer and cytotoxic agents and/or treatments useful in the treatment of cancer or other proliferative diseases are especially useful.
  • anti-cancer and cytotoxic drug combinations wherein the second drug chosen acts in a different manner or different phase of the cell, e.g., S phase, than the present compounds represented by formula I which exert their effects at the G 2 -M phase.
  • Example classes of anti-cancer and cytotoxic agents include, but are not limited to: alkylating agents, such as nitrogen mustards, alkyl sulfonates, nitrosoureas, ethylenimines, and triazenes; antimetabolites, such as folate antagonists, purine analogues, and pyrimidine analogues; antibiotics, such as anthracyclines, bleomycins, mitomycin, dactinomycin, and plicamycin; enzymes, such as L-asparaginase; farnesyl-protein transferase inhibitors; hormonal agents, such as glucocorticoids, estrogens/antiestrogens, androgens/antiandrogens, progestins, and luteinizing hormone-releasing hormone antagonists, octreotide acetate; microtubule-disruptor agents, such as ecteinascidins or their analogs and derivatives; and epothilone
  • the compounds represented by formula I may also be formulated or co-administered with other therapeutic agents that are selected for their particular usefulness in administering therapies associated with the aforementioned conditions.
  • the compounds of the invention may be formulated with agents to prevent nausea, hypersensitivity, and gastric irritation, such as antiemetics, and H 1 and H 2 antihistaminics.
  • the above therapeutic agents when employed in combination with the compounds of the present invention, may be used in those amounts indicated in the Physicians' Desk Reference (PDR) or as otherwise determined by one of ordinary skill in the art.
  • PDR Physicians' Desk Reference
  • the subject compound was administered in either 1:9 ethanol/water, or 1:1:8 Cremphor®/ethanol/water.
  • Final dilution for parenteral administration was made with water one hour before administration.
  • Final dilution for oral administration was made with 0.25 M sodium phosphate buffer.
  • Paclitaxel was dissolved in a 50/50 mixture of ethanol and Cremophor® and maintained at 4° C. Final dilution was made immediately prior to injection to prevent undesirable precipitation.
  • Tumor cell lines HCT 116 human carcinoma and HCT116/V/M46 cells were maintained on McCoy's medium and 10% heat-inactivated fetal bovine serum.
  • A2780 human ovarian carcinoma cells and A2780Tax cells were maintained in IMEM and 10% heat-inactivated fetal bovine serum. All other cell lines were maintained in RPM11640 medium with 10% heat-inactivated fetal bovine serum. Cell lines with acquired resistance will be discussed below.
  • the in vitro cytotoxicity was assessed in tumor cells by a tetrazolium-based colorimetric assay at 492 mm. The cells were seeded 24 h prior to drug addition. The reagents were added following a 72 h incubation with serially diluted test compound. Measurements were taken after a further three hours incubation. The results are expressed as median cytotoxic concentration (IC 50 values).
  • Clonogenic cell colony-formation assay the potency required for the test compound and paclitaxel to kill clonogenic tumor cells (cells that are able to divide indefinitely to form a colony) in vitro was evaluated by a colony formation assay. The concentration needed to kill 90% of clonogenic cancer cells (IC 90 ) was determined.
  • Tubulin polymerization assay the potency required for the test compound and paclitaxel to polymerize tubulin isolated from calf brain was evaluated by published techniques.
  • ovarian carcinoma A2780, A2780Tax and Pat-7 established from an ovarian tumor biopsy from a patient who had developed resistance to paclitaxel
  • HCT116, HCT116/VM46 and LS174T colon carcinomas Pat-21 breast carcinoma, and Pat-26 pancreatic carcinoma (from a liver metastasis biopsy).
  • Pat-7, Pat-21 and Pat-26 xenografts were established initially from primary tumor biopsies directly as xenotransplants grown in whole-body irradiated nude mice without any intervening in vitro cell culturing steps.
  • the innately paclitaxel-insensitive murine fibrosarcoma M5076 was also employed.
  • the human tumor xenografts were maintained in Balb/c nu/nu nude mice.
  • M5076 was maintained in C57BL/6 mice. Tumors were propagated as subcutaneous transplants in the appropriate mouse strain using tumor fragments obtains from donor mice. Tumor passage occupied biweekly for murine tumors and approximately every two to eight weeks for the various human tumor lines.
  • M5076 tumors were implanted in (C57B1/6 ⁇ DBA/2)F1 hybrid mice, and human tumors were implanted in nude mice. All tumor implants for efficacy testing were subcutaneous (sc).
  • the required number of animals needed to detect a meaningful response (6-8) were pooled at the start of the experiment and each was given a subcutaneous implant of a tumor fragment ( ⁇ 50 mg) with a 13-gauge trocar.
  • the animals were again pooled before distribution to the various treatment and control groups.
  • tumors were allowed to grow to the predetermined size window (tumors outside the range were excluded) and animals were evenly distributed to various treatment and control groups.
  • Treatment of each animal was based on individual body weight. Treated animals were checked daily for treatment related toxicity/mortality. Each group of animals was weighed before the inhalation of treatment (Wt1) and then again following the last treatment dose (Wt2). The difference in body weight (Wt2-Wt1) provided a measure of treatment-related toxicity.
  • Tumor response was determined by measurement of tumors with a caliper twice a week until the tumors reached a predetermined “target” size of 0.5 or 1.0 g.
  • the maximum tolerated dose (MTD) is defined as the dose level immediately above which excessive toxicity (i.e. more than one death) occurred.
  • the MTD was frequently equivalent to the optimal dose (OD).
  • Activity is described at the OD.
  • Treated mice expiring prior to having their tumors reach target size were considered to have expired from drug toxicity. No control mice expired bearing tumors less than target size. Treatment groups with more than one death caused by drug toxicity were considered to have had excessively toxic treatments and their data were not included in the evaluation of a compound's antitumor efficacy.
  • Tumor response end-point was expressed in terms of tumor growth delay (T-C value), defined as the difference in time (days) required for the treated tumors (T) to reach a predetermined target size compared to those of the control group (C).
  • T-C value tumor growth delay
  • a tumor is defined as “cured” when there is no detectable disease at the time of study termination; the interval between study termination and the end of drug treatment always exceeded 10 times the tumor volume doubling time.
  • Group sizes typically consisted of eight mice in all treatment and control groups.
  • Statistical analyses of response data were carried out using the Gehan's generalized Wilcoxon test.
  • the results demonstrate the test compound has a broad spectrum of activity against a panel of tumor cell lines in vitro. Of the 21 cells lines tested, the IC 50 values were in the range of 1.4-34.5 nM. Significantly, the test compound appeared to overcome to a large extent the two main mechanisms of resistance to paclitaxel, viz. MDR resistance due to P-glycoprotein overexpression (exemplified by HCT116/VM46) and ⁇ -tubulin mutation (exemplified by A2780Tax). The test compound and paclitaxel were similarly potent in killing clonogenic cells in the two sensitive tumor cell lines (HCT116 and A2780). However, as shown in FIG.
  • test compound performed far better than paclitaxel, almost completely retaining its cytotoxic potency against these resistant cell lines as compared to the sensitive lines.
  • the test compound blocks cells in the mitotic phase of the cell division cycle. Moreover, the concentration of the test compound needed to arrest cells in mitosis, as measured by flow cytometry, corresponds well to the concentration required to kill cells over the same treatment duration. Thus, as shown in FIG. 3 , the test compound at a concentration close to the IC 90 value (about 7.5 nM, clonogenic cytotoxicity assay) almost completely blocks cells in mitosis as early as 8 hours following the initiation of drug exposure.
  • the test compound was evaluated in a panel of eight human and murine tumor models, some of which were chosen because of their known, well-characterized resistance to paclitaxel.
  • the tumor model characteristics are shown in Table 1 below.
  • three paclitaxel-sensitive models were included in order to gain a full assessment of the spectrum of antitumor activity of the test compound.
  • test compound Since the test compound is more stable at neutral pH than at low pH, the evaluation thereof by oral administration (PO) utilized a pH-buffering vehicle (0.25M potassium phosphate, pH 8.0). Using a every 2 days ⁇ 5 schedule, the test compound was highly active orally against the Pat-7 human ovarian carcinoma model. In two separate experiments, orally administered test compound yielded 3.1 and 2.5 LCKs at its MTD. In comparison, concomitantly tested IV paclitaxel produced 1.3 and 1.2 LCK, respectively, at its optimal dose and schedule. Paclitaxel is typically inactive when administered by the oral route.
  • pH-buffering vehicle 0.25M potassium phosphate, pH 8.0
  • test compound retains its antineoplastic activity in cancer cells that have developed resistance to paclitaxel, whether through overexpression of the MDR P-glycoprotein or tubulin mutation. From the in vivo evidence, the test compound has clearly demonstrated antitumor activity superior to paclitaxel in both paclitaxel-resistant and sensitive tumors, and the murine fibrosarcoma M5076. The test compound was more efficacious than paclitaxel in all five paclitaxel-resistant tumors evaluated in this study (four human and one murine); viz.
  • the clinically-derived paclitaxel resistant Pat-7 ovarian carcinoma the A2780Tax ovarian carcinoma that is resistant to paclitaxel because of tubulin mutations
  • the HCT116/VM46 multidrug resistant (MDR) colon carcinoma the clinically-derived paclitaxel-resistant Pat-21 breast carcinoma
  • the murine fibrosarcoma M5076 Against three paclitaxel-sensitive human tumor xenografts, viz. A2780 human ovarians carcinoma; HCT116 and LS 174T human colon carcinoma, the test compound produced antitumor activity equivalent to paclitaxel.
  • test compound over the prototypical taxanes is its efficacy by oral administration, producing antitumor activity when given orally that is equivalent to that produced by IV drug administration in two different human tumor xenografts.

Abstract

Methods of treating tumors in a mammal, especially a human that has demonstrated resistance to other chemotherapeutic agents, is disclosed. Specifically, methods of the present invention are effective in tumors that have initially been unresponsive to taxane therapy, or have developed resistance during the course of treatment. The methods of the present invention comprise administering epothilone derivatives selected from those represented by the formula:
Figure USRE041393-20100622-C00001

The subject epothilone derivatives are advantageous in addition to their enhanced potency and effectiveness against tumors that have demonstrated resistance to therapy with taxane oncology agents in that they are efficacious upon oral administration.

Description

CROSS-REFERENCE TO RELATED APPLICATIONS
This application claims priority from provisional application serial No. 60/269,836, filed Feb. 20, 2001, incorporated herein by reference in its entirety.
FIELD OF THE INVENTION
The present invention relates to the use of certain potent epothilone analogs in the treatment of tumors that have demonstrated resistance to therapy with other chemotherapeutic agents.
BACKGROUND OF THE INVENTION
Epothiolones are macrolide compounds that find utility in the pharmaceutical field. For example, epothilones A and B having the structures:
Figure USRE041393-20100622-C00002

may be found to exert microtubule-stabilizing effects similar to paclitaxel (TAXOL®) and hence cytotoxic activity against rapidly proliferating cells, such as tumor cells or other hyperproliferative cellular diseases. See, Hofle et al., Angew. Chem. Int. Ed. Engl., Vol. 35, No. 13/14, 1567-1569 (1996); WO93/10121 published May 27, 1993; and WO97/19086 published May 29, 1997.
Derivatives and analogs of epothilones A and B have been synthesized and may be used to treat a variety of cancers and other abnormal proliferative diseases. Such analogs are disclosed in Hofle et al., Id.; Nicolaou et al., Agnew. Chem. Int. Ed. Eng., Vol.36, No. 19, 2097-2103 (1997); and Su et al., Agnew. Chem. Int. Ed. Engl., Vol. 36, No. 19, 2093-2097 (1997). In some instances, epothilone derivatives have demonstrated enhanced properties over epothilones A and B. The present invention is concerned with the discovery that certain epothilone derivatives may be utilized to treat cancers that have demonstrated resistance to other chemotherapeutic agents, such as oncolytic agents of the taxane family of compounds.
SUMMARY OF THE INVENTION
In accordance with the present invention, tumors demonstrating a clinical resistance to treatment with other chemotherapeutic agents, such as taxane oncolytic agents, may be treated with an epothilone derivative selected from those represented by formula I:
Figure USRE041393-20100622-C00003

wherein B1, B2, G, Q, X, Y, Z1, Z2, and R1 through R7 have the meanings given below. The compounds represented by formula I have previously demonstrated significantly enhanced potency over known chemotherapeutic agents, for example, epothilones A and B above and certain others including those in the taxane series. Compounds represented by formula I are further advantageous in that, unlike most oncology agents, they are efficacious via oral administration.
BRIEF DESCRIPTION OF THE DRAWINGS
FIG. 1 is a bar graph showing the cytotoxicity spectrum of a compound of the invention against a panel of tumor cell lines.
FIG. 2 is a bar graph showing the cytotoxicity of a compound of the invention against paclitaxel-resistant tumors.
FIG. 3 shows the mitotic blockade induced by a compound of the invention.
DETAILED DESCRIPTION OF THE INVENTION
Processes of the present invention provide advantageous treatment for tumors that have demonstrated resistance to treatment with chemotherapeutic agents, such as those of the taxane family. The term “resistance to treatment” as utilized herein includes both tumors that are initially unresponsive to treatment with a chemotherapeutic agent as well as tumors that are initially responsive, but develop resistance over the course of treatment. Compounds useful in the subject method are epothilones, a class of oncology chemotherapeutic agents chemically distinct from the taxane family of oncology agents. The subject epothilone derivatives are represented by formula I:
Figure USRE041393-20100622-C00004

wherein
    • Q is selected from the group consisting of
      Figure USRE041393-20100622-C00005
    • G is selected from the group consisting of alkyl, substituted alkyl, aryl, substituted aryl, heterocyclo,
      Figure USRE041393-20100622-C00006
    • W is O or N R15;
    • X is O or H, H;
    • Y is selected from the group consisting of O; H, OR16; OR17, OR17; NOR18; H, NHOR19; H, NR20R21; H, H; and CHR25; wherein OR17, OR17 can be cyclic ketal;
    • each Z1 and Z2 is, independently, selected from the group consisting of CH2, O, NR23, S, and SO2, wherein only one of Z1 and Z2 can be a heteroatom;
    • each B1 and B2 is, independently, selected from the group consisting of OR24, OCOR25, and O—C(═O)— NR26R27, and when B1 is H and Y is OH, H, they can form a six-membered ring ketal or acetal;
    • D is selected from the group consisting of NR28R29, NR30COR31 and saturated heterocycle;
    • each R1, R2, R3, R4, R5, R6, R7, R13, R14, R18, R19, R20, R21, R22, R26 and R27 is, independently, selected from the group consisting of H, alkyl, substituted alkyl, and aryl, and when R1 and R2 are alkyl they can be joined to form cycloalkyl, and when R3 and R4 are alkyl they can be joined to form cycloalkyl;
    • each R9, R10, R16, R17, R24, R25 and R31 is, independently, selected from the group consisting of H, alkyl, and substituted alkyl;
    • each R8, R11, R12, R28, R30, R32, and R33 is, independently, selected from the group consisting of H, alkyl, substituted alkyl, aryl, substituted aryl, cycloalkyl and heterocyclo; and
    • each R15, R23 and R29 is, independently, selected from the group consisting of H, alkyl, substituted alkyl, aryl, substituted aryl, cycloalkyl, heterocyclo, R32C═O, R33SO2, hydroxy, O-alkyl or O-substituted alkyl;
      and pharmaceutically acceptable salts thereof and any hydrates, solvates or geometric, optical and stereoisomers thereof,
    • with the proviso that compounds wherein W and X are both O; R1, R2 and R7 are H; R3, R4 and R6 are methyl; R8 is H or methyl; Z1 and Z2 are CH2; G is 1-methyl-2-(substituted-4-thiazolyl)ethenyl; and Q is as defined above, are excluded.
Preferred compounds in accordance with the present invention are those represented by formula I above wherein Q is
Figure USRE041393-20100622-C00007
    • X is O; Y is O; Z1 and Z2 are CH2; and W is NR15.
Another preferred group of compounds in accordance with the present invention is represented below:
  • [1S-[1R*,3R*(E),7R*,10S*,11R*,12R*,16S*]]-7,11-dihydroxy-8,8,10,12,16-pentamethyl-3-[1-methyl-2-(2-methyl-4-thiazolyl)ethenyl]-4,13,17-trioxabicyclo[14.1.0]heptadecane-5,9-dione;
  • [1 S-[1R*,3R*(E),7R*,10S*,11R*,12R*,16S*]]-7,11-dihydroxy-8,8,10,12-tetramethyl-3-[1-methyl-2-(2-methyl-4-thiazolyl)ethenyl]-4,13,17-trioxabicyclo[14.1.0]heptadecane-5,9-dione;
  • [4S-[4R*,7S*,8R*,9R*,15R*(E)]]-4,8-dihydroxy-5,5,7,9,13-pentamethyl-16-[1-methyl-2-(2-methyl-4-thiazolyl)ethenyl]-1,10-dioxa-13-cyclohexadecene-2,6-dione;
  • [4S-[4R*,7S *,8R*,9R*,15R*(E)]]-4,8-dihydroxy-5,5,7,9-tetramethyl-16-[1-methyl-2-(2-methyl-4-thiazolyl)ethenyl]-1,10-dioxa-13-cyclohexadecene-2,6-dione;
  • [1S-[1R*,3R*(E),7R*,10S*,11R*,12R*,16S*]]-7,11-dihydroxy-8,8,10,12,16-pentamethyl-3-[1-methyl-2-(2-methyl-4-thiazolyl)ethenyl]-4,14,17-trioxabicyclo[14.1.0]heptadecane-5,9-dione;
  • [1S-[1R*,3R*(E),7R*,10S*,11R*,12R*,16S*]]-7,11-dihydroxy-8,8,10,12-tetramethyl-3-[1-methyl-2-(2-methyl-4-thiazolyl)ethenyl]-4,14,17-trioxabicyclo[14.1.0]heptadecane-5,9-dione;
  • [4S-[4R*,7S*,8R*,9R*,15R*(E)]]-4,8-dihydroxy-5,5,7,9,13-pentamethyl-16-[1-methyl-2-(2-methyl-4-thiazolyl)ethenyl]-1,11-dioxa-13-cyclohexadecene-2,6-dione;
  • [4S-[4R*,7S*,8R*,9R*,15R*(E)]]-4,8-dihydroxy-5,5,7,9-tetramethyl-16-[1-methyl-2-(2-methyl-4-thiazolyl)ethenyl]-1,11-dioxa-13-cyclohexadecene-2,6-dione;
  • [1S-[1R*,3R*(E),7R*,10S*,11R*,12R*,16S*]]-7,11-dihydroxy-8,8,10,12,16-pentamethyl-3-[1-methyl-2-(2-methyl-4-thiazolyl)ethenyl]-4,17-dioxabicyclo[14.1.0]heptadecane-9-one;
  • [1S-[1R*,3R*(E),7R*,10S*,11R*,12R*,16S*]]-7,11-dihydroxy-8,8,10,12-tetramethyl-3-[1-methyl-2-(2-methyl-4-thiazolyl)ethenyl]-4,17-dioxabicyclo[14.1.0]heptadecane-9-one; hexamethyl-3-[1-methyl-2-(2-methyl-4-thiazolyl)ethenyl]-4, 17-dioxabicyclo[14.1.0]heptadecane-5,9-dione;
  • [1S-[1R*,3R*(E),7R*,10S*,11R*,12R*,16S*]]-7,11-dihydroxy-3,8,8,10,12-pentamethyl-3-[1-methyl-2-(2-methyl-4-thiazolyl)ethenyl]-4,17-dioxabicyclo[14.1.0]heptadecane-5,9-dione;
  • [4S-[4R*,7S*,8R*,9R*,15R*(E)]]-4,8-dihydroxy-5,5,7,9,13,16-hexamethyl-16-[1-methyl-2-(2-methyl-4-thiazolyl)ethenyl]-1-oxa-13-cyclohexadecene-2,6-dione;
  • [4S-[4R*,7S*,8R*,9R*,15R*(E)]]-4,8-dihydroxy-5,5,7,9,16-pentamethyl-16-[1-methyl-2-(2-methyl-4-thiazolyl)ethenyl]-1-oxa-13-cyclohexadecene-2,6-dione;
  • [1 S-[R*,3R*(E),7R*,10S*,11R*,12R*,16S*]]-7,11-dihydroxy-8,8,10,12,16-pentamethyl-3-[1-methyl-2-(2-methyl-4-thiazolyl)ethenyl]-4,17-dioxabicyclo[14.1.0]heptadecane-5,9-dione;
  • [1S-[1R*,3R*(E),7R*,10S*,11 R*,12R*,16S*]]-7,11-dihydroxy-6,8,8,10,12-pentamethyl-3-[1-methyl-2-(2-methyl-4-thiazolyl)ethenyl]-4,17-dioxabicyclo[14.1.0]heptadecane-5,9-dione;
  • [1S-[1R*,3R*(E),7R*,10S*,11R*,12R*,16S*]]-7,11-dihydroxy-8,8,10,12,16-pentamethyl-3-[1-methyl-2-(2-methyl-4-thiazolyl)ethenyl]-4-aza-17-oxabicyclo[14.1.0]heptadecane-5,9-dione;
  • [1 S-[1R*,3R*(E),7R*,10S*,11R*,12R*,16S*]]-7,11-dihydroxy-8,8,10,12-tetramethyl-3-[1-methyl-2-(2-methyl-4-thiazolyl)ethenyl]-4-aza-17-oxabicyclo[14.1.0]heptadecane-5,9-dione;
  • [4S-[4R*,7S *,8R*,9R*,15R*(E)]]-4,8-dihydroxy-5,5,7,9,13-pentamethyl-16-[1-methyl-2-(2-methyl-4-thiazolyl)ethenyl]-1-aza-13-cyclohexadecene-2,6-dione;
  • [4S-[4R*,7S*,8R*,9R*,15R*(E)]]-4,8-dihydroxy-5,5,7,9-tetramethyl-16-[1-methyl-2-(2-methyl-4-thiazolyl)ethenyl]-1-aza-13-cyclohexadecene-2,6-dione;
  • [1S-[1R*,3R*(E),7R*,10S*,11R*,12R*,16S*]]-7,11-dihydroxy-4,8,8,10,12,16-hexamethyl-3-[1-methyl-2-(2-methyl-4-thiazolyl)ethenyl]-4-aza-17-oxabicyclo[14.1.0]heptadecane-5,9-dione;
  • [1S-[R*,3R*(E),7R*,10S*,11R*,12R*,16S*]]-7,11-dihydroxy-4,8,8,10,12-pentamethyl-3-[1-methyl-2-(2-methyl-4-thiazolyl)ethenyl]-4-aza-17-oxabicyclo[14.1.0]heptadecane-5,9-dione;
  • [4S-[4R*,7S*,8R*,9 R*,15R*(E)4,8-dihydroxy-1,5,5,7,9,13-hexamethyl-16-[1-methyl-2-(2-methyl-4-thiazolyl)ethenyl]-1-aza-13-cyclohexadecene-2,6-dione;
  • [4S-[4R*,7S*,8R*,9R*,15R*(E)]]-4,8-dihydroxy-1,5,5,7,9-pentamethyl-16-[1-methyl-2-(2-methyl-4-thiazolyl)ethenyl]-1-aza-13-cyclohexadecene-2,6-dione;
  • [1S-[1R*,3R*(E),7R*,10S*,11R*,12R*,16S*]]-7,11-dihdyroxy-8,8,10,12,16-pentamethyl-3-[1-methyl-2-(2-methyl-4-thiazolyl)ethenyl]-13-aza-4,17-dioxabicyclo[14.1.0]heptadecane-5,9-dione;
  • [1 S-[1R*,3R*(E),7R*,10S*,11R*,12R*,16S*]]-7,11-dihydroxy-8,8,10,12-tetramethyl-3-[1-methyl-2-(2-methyl-4-thiazolyl)ethenyl]-13-aza-4,17dioxabicyclo[14.1.0]heptadecane-5,9-dione;
  • [4S-[4R*,7S*,8R*,9R*,15R*(E)]]-4,8-dihydroxy-5,5,7,9,13-pentamethyl-16-[1-methyl-2-(2-methyl-4-thiazolyl)ethenyl]-10-aza-1-oxa-13-cyclohexadecene-2,6-dione;
  • [4S-[4R*,7S*,8R*,9R*,15R*(E)]]-4,8-dihydroxy-5,5,7,9-tetramethyl-16-[1-methyl-2-(2-methyl-4-thiazolyl)ethenyl]-10-aza-1-oxa-13-cyclohexadecene-2,6-dione;
  • [1S-[1R*,3R*(E),7R*,10S*,11R*,12R*,16S*]]-7,11-dihydroxy-8,8,10,12,16-pentamethyl-3-[1-methyl-2-(2-methyl-4-thiazolyl)ethenyl]-14-aza-4,17-dioxabicyclo[14.1.0]heptadecane-5,9-dione;
  • [1S-[R*,3R*(E),7R*,10S*,11R*,12R*,16S* ]]-7,11-dihydroxy-8,8,10,12-tetramethyl-3-[1-methyl-2-(2-methyl-4-thiazolyl)ethenyl]-14-aza-4,17-dioxabicyclo[14.1.0]heptadecane-5,9-dione;
  • [4S-[4R*,7S,8R*,9R*,15R*(E)]]-4,8-dihydroxy-5,5,7,9,13-pentamethyl-16-[1-methyl-2-(2-methyl-4-triazolyl)ethenyl]-11-aza-1-oxa-13-cyclohexadecene-2,6-dione;
  • [4S-[4R*,7S*,8R*,9R*15R*(E)]]-4,8-dihydroxy-5,5,7,9-tetramethyl-16-[1-methyl-2-(2-methyl-4-thiazolyl)ethenyl]-11-aza-1-oxa-13-cyclohexadecene-2,6-dione;
  • [1 S-[1R*,3R*,7R*,10S*,11R*,12R*,16S*]]-N-phenyl-7,11-dihydroxy-8,8,10,12,16-pentamethyl-5,9-dioxo-4,17-dioxabicyclo[14.1.0]heptadecane-3-carboxamide;
  • [1S-[1R*,3R*,7R*,10S*,11R*,12R*,16S*]]-N-phenyl-7,11-dihydroxy-8,8,10,12-tetramethyl-5,9-dioxo-4,17-dioxabicyclo[14.1.0]heptadecane-3-carboxamide;
  • 4S-[4R*,7S *,8R*,9R*,15R*]]-N-phenyl-4,8-dihydroxy-5,5,7,9,13-pentamethyl-2,6-dioxo-1-oxa-13-cyclohexadecene-16-carboxamide;
  • [4S-[4R*,7S,8R*,9R*,15R*]]-N-phenyl-4,8-dihydroxy-5,5,7,9-tetramethyl-2,6-dioxo-1-oxa-13-cyclohexadecene-16-carboxamide;
    • [1S-1R*,3R*(E),7R*,10S*,11R*,12R*,16S*]]-7,11-dihydroxy-8,8,10,12,16-pentamethyl-3-[1-methyl-2-(2-methyl-4-thiazolyl)cyclopropyl]-4,17-dioxabicyclo[14.1.0]heptadecane-5,9-dione;
  • 1[S-[1R*,3R*(E),7R*,10S*,11R, 12R*,16S*]]-7,11-dihydroxy-8,8,10,12-tetramethyl-3-[1-methyl-2-(2-methyl-4-thiazolyl)cyclopropyl]-4,17-dioxabicyclo[14.1.0]heptadecane-5,9-dione; and
  • [4S-[4R*,7S*,8R*,9R*,15R*(E)]]-4,8-dihydroxy-5,5,7,9,13-pentamethyl-16-[1-methyl-2-(2-hydroxymethyl-4-thiazolyl)ethenyl]-1-aza-13(Z)-cyclohexadecene-2,6-dione;
    and pharmaceutically acceptable salts, solvates and hydrates thereof.
A particular preferred compound in accordance with the present invention is represented by the formula:
Figure USRE041393-20100622-C00008

This compound chemically is [1S-[1R*,3R*(E),7R*,10S*,11R*,12R*,16S*]]-7,11-dihydroxy-8,8,10,12,16-pentamethyl-3-[1-methyl-2-(2-methyl-4-thiazolyl)ethenyl]-4-aza-17-oxabicyclo[14.1.0]heptadecane-5,9-dione.
The epothilone derivatives represented by formula I above and processes for their preparation are disclosed in WO 99/02514, WO 99/27890, WO 99/28324. Heretofore, however, there has been no recognition that the subject epothilone derivatives would possess activity in the treatment of tumors resistant to treatment with other known chemotherapeutic agents.
The following are definitions of various terms used to describe the compound represented by formula I above.
The term “alkyl” refers to optionally substituted straight- or branched-chain saturated hydrocarbon groups having from 1 to about 20 carbon atoms, preferably from 1 to about 7 carbon atoms. The expression “lower alkyl” refers to optionally substituted alkyl groups having from 1 to about 4 carbon atoms.
The term “substituted alkyl” refers to an alkyl group substituted by, for example, one to four substituents, such as, halo, trifluoromethyl, trifluoromethoxy, hydroxy, alkoxy, cycloalkyoxy, heterocylooxy, oxo, alkanoyl, aryl, aryloxy, aralkyl, alkanoyloxy, amino, alkylamino, arylamino, aralkylamino, cycloalkylamino, heterocycloamino, disubstituted amino in which the two substituents on the amino group are selected from alkyl, aryl, aralkyl, alkanoylamino, aroylamino, aralkanoylamino, substituted alkanoylamino, substituted arylamino, substituted aralkanoylamino, thiol, alkylthio, arylthio, aralkylthio, cycloalkylthio, heterocyclothio, alkylthiono, arylthiono, aralkylthiono, alkylsulfonyl, arylsulfonyl, aralkylsulfonyl, sulfonamido (e.g., SO2NH2), substituted sulfonamido, nitro, cyano, carboxy, carbamyl (e.g., CONH2), substituted carbamyl (e.g., CONH alkyl, CONH aryl, CONH aralkyl or instances where there are two substituents on the nitrogen selected from alkyl, aryl or aralkyl), alkoxycarbonyl, aryl, substituted aryl, guanidino and heterocyclos, such as, indolyl, imidazolyl, furyl, thienyl, thiazolyl, pyrrolidyl, pyridyl, pyrimidyl and the like. Wherein, as noted above, the substituents themselves are further substituted, such further substituents are selected from the group consisting of halogen, alkyl, alkoxy, aryl and aralkyl. The definitions given herein for alkyl and substituted alkyl apply as well to the alkyl portion of alkoxy groups.
The term “alkenyl” refers to optionally substituted unsaturated aliphatic hydrocarbon groups having from 1 to about 9 carbons and one or more double bonds. Substituents may include one or more substituent groups as described above for substituted alkyl.
The term “halogen” or “halo” refers to fluorine, chlorine, bromine and iodine.
The term “ring system” refers to an optionally substituted ring system containing one to three rings and at least one carbon to carbon double bond in at least one ring. Exemplary ring systems include, but are not limited to, an aryl or a partially or fully unsaturated heterocyclic ring system, which may be optionally substituted.
The term “aryl” refers to monocyclic or bicyclic aromatic hydrocarbon groups having from about 6 to about 12 carbon atoms in the ring portion, for example, phenyl, naphthyl, biphenyl and diphenyl groups, each of which may be substituted.
The term “aralkyl” refers to an aryl group bonded to a larger entity through an alkyl group, such as benzyl.
The term “substituted aryl” refers to an aryl group substituted by, for example, one to four substituents such as alkyl; substituted alkyl, halo, trifluoromethyl, trifluoromethoxy, hydroxy, alkoxy, cycloalkyloxy, heterocyclooxy, alkanoyl, alkanyloxy, amino, alkylamino, dialkylamino, aralkylamino, cycloalkylamino, heterocycloamino, alkanoylamino, thiol, alkylthio, cycloalkylthio, heterocyclothio, ureido, nitro, cyano, carboxy, carboxyalkyl, carbamyl, alkoxycarbonyl, alkylthio, arylthiono, alkylsulfonyl, sulfonamido, aryloxy and the like. The substituent may be further substituted by one or more members selected from the group consisting of halo, hydroxy, alkyl, alkoxy, aryl, substituted alkyl, substituted aryl and aralkyl.
The term “cycloalkyl” refers to optionally substituted saturated cyclic hydrocarbon ring systems, preferably containing 1 to about 3 rings and 3 to about 7 carbon atoms per ring, which may be further fused with an unsaturated C3-C7 carbocyclic ring. Exemplary groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, cyclodecyl, cyclododecyl, and adamantyl. Exemplary substituents include one or more alkyl groups as described above, or one or more of the groups described above as substituents for alkyl groups.
The terms “heterocycle”, “heterocyclic” and “heterocyclo” refer to an optionally substituted, unsaturated, partially saturated, or fully saturated, aromatic or nanoaromatic cyclic group, for example, which is a 4- to 7-membered monocyclic, 7-to 11-membered bicyclic, or 10- to 15-membered tricyclic ring system, which has at least one heteroatom in at least one carbon atom-containing ring. Each ring of the heterocyclic group containing a heteroatom may have 1, 2 or 3 heteroatoms selected from nitrogen atoms, oxygen atoms and sulfur atoms, where the nitrogen and sulfur heteroatoms may also optionally be oxidized and the nitrogen heteroatoms may also optionally be quaternized. The heterocyclic group may be attached at any heteroatom or carbon atom.
Exemplary monocyclic heterocyclic groups include pyrrolidinyl, pyrrolyl, indolyl, pyrazolyl, oxetanyl, pyrazolinyl, imidazolyl, imidazolinyl, imidazolidinyl, oxazolyl, oxazolidinyl, isoxazolinyl, isoxazolyl, thiazolyl, thiadiazolyl, thiazolidinyl, isothiazolyl, isothiazolidinyl, furyl, tetrahydrofuryl, thienyl, oxadiazolyl, piperidinyl, piperizinyl, 2-oxopiperazinyl, 2-oxopiperidinyl, 2-oxopyrrolidinyl, 2-oxazepinyl, azepinyl, 4-piperidonyl, pyridyl, N-oxo-pyridyl, pyrazinyl, pyrimidinyl, pyridazinyl, tetrahydropyranyl, tetrahydrothiopyranyl, tetrahydrothiopyranyl sulfone, morpholinyl, thiomorpholinyl, thiomorpholinyl sulfoxide, thiomorpholinyl sulfone, 1,3-dioxolane and tetrahydro-1,1-dioxothienyl, dioxanyl, isothiazolidinyl, thietanyl, thiiranyl, triazinyl, and triazolyl, and the like.
Exemplary bicyclic heterocyclic groups include benzothiazolyl, benzoxazolyl, benzothienyl, quinuclidinyl, quinolinyl, quinolinyl-N-oxide, tetrahydroisoquinolinyl, isoquinolinyl, benzimidazolyl, benzopyranyl, indolizinyl, benzofuryl, chromonyl, coumarinyl, cinnolinyl, quinoxalinyl, indazolyl, pyrrolopyridyl, furopyridinyl (such as furo[2,3-c]pyridinyl, furo[3,1-b]pyridinyl] or furo[2,3-b]pyridinyl), dihydroisoindolyl, dihydroquinazolinyl (such as 3,4-dihydro-4-oxo-quinazolinyl), benzisothiazolyl, benzisoxazolyl, benzodiazinyl, benzofurazanyl, benzothiopyranyl, benzotriazolyl, benzpyrazolyl, dihydrobenzofuryl, dihydrobenzothienyl, dihydrobenzothiopyranyl, dihydrobenzothiopyranyl sulfone, dihydrobenzopyranyl, indolinyl, isochromanyl, isoindolinyl, naphthyridinyl, phthalazinyl, piperonyl, purinyl, pyridopyridyl, quinazolinyl, tetrahydroquinolinyl, thienofuryl, thienopyridyl, thienothienyl, and the like.
Exemplary substituents for the terms “ring system,” “heterocycle,” “heterocyclic,” and “heterocyclo” include one or more substituent groups as described above for substituted alkyl or substituted aryl, and smaller heterocycles, such as, epoxides, aziridines and the like.
The term “alkanoyl” refers to —C(O)-alkyl.
The term “substituted alkanoyl” refers to —C(O)-substituted alkyl.
The term “heteroatoms” shall include oxygen, sulfur and nitrogen.
The compounds represented by formula I form salts with a variety of organic and inorganic acids. Such salts include those formed with hydrogen chloride, hydrogen bromide, methanesulfonic acid, hydroxyethanesulfonic acid, sulfuric acid, acetic acid, trifluoroacetic acid, maleic acid, benzenesulfonic acid, toluenesulfonic acid and various others as are recognized by those of ordinary skill in the art of pharmaceutical compounding. Such salts are formed by reacting a compound represented by formula I in an equivalent amount of the acid in a medium in which the salt precipitates or in an aqueous medium followed by evaporation.
In addition, zwitterions (“inner salts”) can be formed and are included within the term “salts” as used herein. Further, solvates and hydrates of the compounds represented by formula I are also included herein.
The compounds represented by formula I above may exist as multiple optical, geometric, and stereoisomers. While the compounds shown herein are depicted for one optical orientation, included within the present invention are all isomers and mixtures thereof.
It is recognized that the compounds represented by formula I above are microtubule-stabilizing agents. Therefore, they are useful in the treatment of a variety of cancers and other proliferative diseases including, but not limited to, the following:
    • carcinoma, including that of the bladder, breast, colon, kidney, liver, lung, ovary, pancreas, stomach, cervix, thyroid and skin, including aquamous cell carcinoma;
    • hematopoietic tumors of lymphoid lineage, including leukemia, acute lymphocytic leukemia, acute lymphoblastic leukemia, B-cell lymphoma, T-cell lymphoma, Hodgkins lymphoma, non-Hodgkins lympoma, hairy cell lymphoma and Burketts lymphoma;
    • hematopoietic tumors of meyloid lineage, including acute and chronic myelogenous leukemias and promyelocytic lumekia;
    • tumors of mesenchymal origin, including fibrosarcoma and rhabdomyosarcoma;
    • other tumors, including melanoma, seminoma, teratocarcinoma, neuroblastoma and glioma;
    • tumors of the central and peripheral nervous system, including astrocytoma, neuroblastoma, glioma, and schwannomas;
    • tumors of mesenchymal origin, including fibrosarcoma, rhabdomyosarcoma, and osteosarcoma; and
    • other tumors, including melanoma, xeroderma pigmentosum, keratoacanthoma, seminoma, thyroid follicular cancer and teratocarcincoma.
The foregoing indications are given herein since it cannot be certain which of the named types of tumors, and others as well, may demonstrate resistance to oncology therapy. “Oncology therapy” refers to treatment of cancer or tumors with chemotherapeutic agents that exert a cytotoxic effect in cells. An example of a chemotherapeutic agent is an oncology agent of the taxane family of compounds. It is known, for example, that a considerable number of patients initially responsive to oncology therapy with taxane compounds develop resistance over a course of therapy and that not all cancers respond to treatment with taxane therapy as is the case with virtually all oncology agents. Further, certain diseases, such as cholorectal cancers or melanoma, are known to be innately resistant to taxane therapy.
The subject epothilone compounds are highly potent cytotoxic agents capable of killing cancer cells at low nanometer concentrations and are approximately twice as potent as paclitaxel in inducing tubulin polymerization. More important, the subject compound seem to possess the capacity to retain their antineoplastic activity against human cancers that are naturally insensitive to paclitaxel or have developed resistance to it, both in vitro and in vivo.
Tumors for which the subject epothilone compounds have demonstrated significant antitumor activity include, without intended limitation, the following:
    • [1] Paclitaxel-resistant—HCT 116VM46 colorectal (multidrug resistant, MDR), Pat-21, breast and Pat-7 ovarian carcinoma (clinical isolates, mechanisms of resistance not fully known), A2780Tax ovarian carcinoma (tubulin mutation);
    • [2] Paclitaxel-insensitive—Pat-26 human pancreatic carcinoma (clinical isolate) and M5076 murine fibrosarcoma; and
    • [3] Paclitaxel sensitive—A2780 ovarian, LS 174T and HCT human colon carcinoma.
In addition, the compounds represented by formula I have demonstrated that they are orally efficacious versus preclinical human tumor xenografts grown in immunocompromized mice or rats. Being efficacious upon oral administration is considered a significant advantage of the subject epothilone derivatives.
The present invention thus provides a method of treating a subject, preferably mammals and especially humans, in need of treatment for a tumor that has demonstrated resistance to therapy with the taxane family of oncologic agents, comprising administering to the subject one of the epothilone compounds represented by formula I in an amount effective for such treatment. Other therapeutic agents, such as those described below, may be employed with the subject epothilone compounds in their usual dosages. Such agents may be administered prior to, simultaneously with, or following the subject epothilone compounds.
An effective amount of the epothilone compounds represented by formula I may be determined by one of ordinary skill in the art, and includes exemplary dosage amounts for a human of from about 0.05 to about 200 mg/kg/day. This dosage is typically administered in a single dose, but can be administered in divided doses since the subject compounds are advantageously efficacious via oral administration. The compounds may be administered in a frequent regimen, e.g., every two days for five doses, or intermittently, e.g., every four days for three doses or every eight days for three doses. It will be understood that the specific dose level and frequency of administration for a given subject may be varied and will depend upon a variety of factors, including the subject's age, body weight, general health, sex, diet and the like, the mode of administration if not oral, severity of the condition and the like.
The compounds represented by formula I are administered in pharmaceutical compositions containing an amount thereof effective for cancer therapy, and a pharmaceutically acceptable carrier. Such compositions may contain other therapeutic agents as described below, and may be formulated, for example, by employing conventional solid or liquid vehicles or diluents, as well as pharmaceutical additives of a type appropriate to the mode of desired administration (for example, excipients, binders, preservatives, stabilizers, flavors, etc.) according to techniques such as those well known in the art of pharmaceutical formulation and/or called for accepted pharmaceutical practice.
The compounds represented by formula I may be administered by any suitable means, for example, orally, such as in the form of tablets, capsules, granules or powders; sublingually; bucally; parenterally, such as by subcutaneous, intravenous, intramuscular, or intrasternal injection or infusion techniques (e.g., as sterile injectable aqueous or non-aqueous solutions or suspensions); nasally, such as by inhalation spray; topically, such as in the form of a cream or ointment; or rectally such as in the form of suppositories; in dosage unit formulations containing non-toxic, pharmaceutically acceptable vehicles or diluents. The subject compounds may, for example, be administered in a form suitable for immediate release or extended release. Immediate release or extended release may be achieved by the use of suitable pharmaceutical compositions comprising the present compounds, or, particularly in the case of extended release, by the use of devices such as subcutaneous implants or osmotic pumps. The subject compounds may also be administered liposomally.
Suitable dosage forms for the subject epothilone derivatives include, without intended limitation, a orally effective composition such as a tablet, capsule, solution or suspension containing about 5 to about 500 mg per unit dosage of a compound represented by formula I or a topical form (about 0.01% to about 5% by weight compound represented by formula I, one to five treatments per day). They may be compounded in a conventional manner with a physiologically acceptable vehicle or carrier, excipient, binder, preservative, stabilizer, flavor, etc., or with a topical carrier. The compounds represented by formula I can also be formulated in compositions such as sterile solutions or suspensions for parenteral administration. About 0.1 mg to about 500 mg of a compound represented by formula I may be compounded with a physiologically acceptable vehicle, carrier, excipient, binder preservative, stabilizer, etc., in a unit dosage form as called for by accepted pharmaceutical practice. The amount of active substance in these compositions or preparations is preferably such that a suitable dosage in the range indicated is obtained.
Exemplary compositions for oral administration include suspensions which may contain, for example, microcrystalline cellulose for imparting bulk, alginic acid or sodium alginate as a suspending agent, methylcellulose as a viscosity enhancer, and sweeteners, or flavoring agents such as those known in the art; and immediate release tablets which may contain, for example, microcrystalline cellulose, dicalcium phosphate, starch, magnesium stearate and/or lactose and/or other excipients, binders, extenders, disintegrants, diluents and lubricants such as those known in the art. Molded tablets, compressed tablets or freeze-dried tablets are exemplary forms that may be used. Exemplary compositions include those formulating the present compound(s) with fast dissolving diluents such as mannitol, lactose, sucrose and/or cyclodextrins. Also included in such formulations may be high molecular weight excipients such as celluloses (Avicel) or polyethylene glycols (PEG). Such formulations may also include an excipient to aid mucosal adhesion such as hydroxy propyl cellulose (HPC), hydroxy propyl methyl cellulose (HPMC), sodium carboxy methyl cellulose (SCMC), maleic anhydride copolymer (e.g. Gantrez), and agents to control release such as polyacrylic acid copolymer (e.g. Carbopol 934). Lubricants, glidants, flavors, coloring agents and stabilizers may also be added for ease of fabrication and use.
Exemplary compositions for nasal aerosol or inhalation administration include solutions in saline which may contain, for example, benzyl alcohol or other suitable preservatives, absorption promoters to enhance bioavailability, and/or other solubilizing or dispersing agents such as those known in the art.
Exemplary compositions for parenteral administration include injectable solutions or suspensions which may contain, for example, suitable non-toxic, parentally acceptable diluents or solvents, such as Cremophor® (polyoxyethylated caster oil surfactant), mannitol, 1,3-butanediol, water, Ringer's solution, Lactated Ringer's solution, an isotonic sodium chloride solution, or other suitable dispersion or wetting and suspending agents, including synthetic mono- or diglycerides, and fatty acids, including oleic acid. Exemplary compositions for rectal administration include suppositories which may contain, for example, a suitable non-irritating excipient, such as cocoa butter, synthetic glyceride esters or polyethylene glycols, which are solid at ordinary temperature, but liquefy and/or dissolve in the rectal cavity to release the drug.
The compounds of the invention may be administered either alone or in combination with other chemotherapeutic agents or anti-cancer and cytotoxic agents and/or treatments useful in the treatment of cancer or other proliferative diseases. Especially useful are anti-cancer and cytotoxic drug combinations wherein the second drug chosen acts in a different manner or different phase of the cell, e.g., S phase, than the present compounds represented by formula I which exert their effects at the G2-M phase. Example classes of anti-cancer and cytotoxic agents include, but are not limited to: alkylating agents, such as nitrogen mustards, alkyl sulfonates, nitrosoureas, ethylenimines, and triazenes; antimetabolites, such as folate antagonists, purine analogues, and pyrimidine analogues; antibiotics, such as anthracyclines, bleomycins, mitomycin, dactinomycin, and plicamycin; enzymes, such as L-asparaginase; farnesyl-protein transferase inhibitors; hormonal agents, such as glucocorticoids, estrogens/antiestrogens, androgens/antiandrogens, progestins, and luteinizing hormone-releasing hormone antagonists, octreotide acetate; microtubule-disruptor agents, such as ecteinascidins or their analogs and derivatives; and epothilones A-F or their analogs or derivatives; plant-derived products, such as vinca alkaloids, epipodophyllotoxins, and topoisomerase inhibitors; prenyl-protein transferse inhibitors; and miscellaneous agents such as, hydroxyurea, procarbazine, mitotane, hexamethylmelarnine, platinum coordintination complexes such as cisplatin and carboplatin; and other agents used as anti-cancer and cytotoxic agents such as biological response modifiers, growth factors; immune modulators, and monoclonal antibodies. The subject compounds may also be used in conjunction with radiation therapy.
The compounds represented by formula I may also be formulated or co-administered with other therapeutic agents that are selected for their particular usefulness in administering therapies associated with the aforementioned conditions. For example, the compounds of the invention may be formulated with agents to prevent nausea, hypersensitivity, and gastric irritation, such as antiemetics, and H1 and H2 antihistaminics.
The above therapeutic agents, when employed in combination with the compounds of the present invention, may be used in those amounts indicated in the Physicians' Desk Reference (PDR) or as otherwise determined by one of ordinary skill in the art.
The following example is provided, without any intended limitation, to further illustrate the present invention.
EXAMPLE
[1 S-[1R*,3R*(E),7R*,10S*,11R*,12R*,16S*]]-7,11-1-Dihydroxy-8,8,10,12,16-pentamethyl-3-[1-methyl-2-(2-methyl-4-thiazolyl)ethenyl]-4-aza-17-oxabicyclo[14.1.0]heptadecane-5,9-dione (BMS-247550).
For administration to rodents, the subject compound was administered in either 1:9 ethanol/water, or 1:1:8 Cremphor®/ethanol/water. Final dilution for parenteral administration was made with water one hour before administration. Final dilution for oral administration was made with 0.25 M sodium phosphate buffer. Paclitaxel was dissolved in a 50/50 mixture of ethanol and Cremophor® and maintained at 4° C. Final dilution was made immediately prior to injection to prevent undesirable precipitation. Tumor cell lines HCT 116 human carcinoma and HCT116/V/M46 cells were maintained on McCoy's medium and 10% heat-inactivated fetal bovine serum. A2780 human ovarian carcinoma cells and A2780Tax cells were maintained in IMEM and 10% heat-inactivated fetal bovine serum. All other cell lines were maintained in RPM11640 medium with 10% heat-inactivated fetal bovine serum. Cell lines with acquired resistance will be discussed below.
The in vitro cytotoxicity was assessed in tumor cells by a tetrazolium-based colorimetric assay at 492 mm. The cells were seeded 24 h prior to drug addition. The reagents were added following a 72 h incubation with serially diluted test compound. Measurements were taken after a further three hours incubation. The results are expressed as median cytotoxic concentration (IC50 values).
Clonogenic cell colony-formation assay: the potency required for the test compound and paclitaxel to kill clonogenic tumor cells (cells that are able to divide indefinitely to form a colony) in vitro was evaluated by a colony formation assay. The concentration needed to kill 90% of clonogenic cancer cells (IC90) was determined.
Tubulin polymerization assay: the potency required for the test compound and paclitaxel to polymerize tubulin isolated from calf brain was evaluated by published techniques. The effective concentration (EC0.01) was defined as the interpolated concentration capable of inducing an initial slope of optical density (OD) of 0.01 OD/minute rate and is calculated using the formula: EC0.01=concentration/slope. EC0.01 values are expressed as the mean with standard deviation obtained from 3 different concentrations.
In Vivo Antitumor Testing: The following human tumors were used: ovarian carcinoma A2780, A2780Tax and Pat-7 (established from an ovarian tumor biopsy from a patient who had developed resistance to paclitaxel); HCT116, HCT116/VM46 and LS174T colon carcinomas, Pat-21 breast carcinoma, and Pat-26 pancreatic carcinoma (from a liver metastasis biopsy). Pat-7, Pat-21 and Pat-26 xenografts were established initially from primary tumor biopsies directly as xenotransplants grown in whole-body irradiated nude mice without any intervening in vitro cell culturing steps. The innately paclitaxel-insensitive murine fibrosarcoma M5076 was also employed. The human tumor xenografts were maintained in Balb/c nu/nu nude mice. M5076 was maintained in C57BL/6 mice. Tumors were propagated as subcutaneous transplants in the appropriate mouse strain using tumor fragments obtains from donor mice. Tumor passage occupied biweekly for murine tumors and approximately every two to eight weeks for the various human tumor lines. With regard to efficacy testing, M5076 tumors were implanted in (C57B1/6×DBA/2)F1 hybrid mice, and human tumors were implanted in nude mice. All tumor implants for efficacy testing were subcutaneous (sc).
The required number of animals needed to detect a meaningful response (6-8) were pooled at the start of the experiment and each was given a subcutaneous implant of a tumor fragment (˜50 mg) with a 13-gauge trocar. For treatment of early-stage tumors, the animals were again pooled before distribution to the various treatment and control groups. For treatment of animals with advanced-stage disease, tumors were allowed to grow to the predetermined size window (tumors outside the range were excluded) and animals were evenly distributed to various treatment and control groups. Treatment of each animal was based on individual body weight. Treated animals were checked daily for treatment related toxicity/mortality. Each group of animals was weighed before the inhalation of treatment (Wt1) and then again following the last treatment dose (Wt2). The difference in body weight (Wt2-Wt1) provided a measure of treatment-related toxicity.
Tumor response was determined by measurement of tumors with a caliper twice a week until the tumors reached a predetermined “target” size of 0.5 or 1.0 g. Tumor weights (mg) were estimated from the formula:
Tumor weight=(length×weight)÷2
The maximum tolerated dose (MTD) is defined as the dose level immediately above which excessive toxicity (i.e. more than one death) occurred. The MTD was frequently equivalent to the optimal dose (OD). Activity is described at the OD. Treated mice expiring prior to having their tumors reach target size were considered to have expired from drug toxicity. No control mice expired bearing tumors less than target size. Treatment groups with more than one death caused by drug toxicity were considered to have had excessively toxic treatments and their data were not included in the evaluation of a compound's antitumor efficacy.
Tumor response end-point was expressed in terms of tumor growth delay (T-C value), defined as the difference in time (days) required for the treated tumors (T) to reach a predetermined target size compared to those of the control group (C). A tumor is defined as “cured” when there is no detectable disease at the time of study termination; the interval between study termination and the end of drug treatment always exceeded 10 times the tumor volume doubling time. Group sizes typically consisted of eight mice in all treatment and control groups. Statistical analyses of response data were carried out using the Gehan's generalized Wilcoxon test.
Cytotoxicity Against Cancer Cells in vitro: As shown in FIG. 1, the results demonstrate the test compound has a broad spectrum of activity against a panel of tumor cell lines in vitro. Of the 21 cells lines tested, the IC50 values were in the range of 1.4-34.5 nM. Significantly, the test compound appeared to overcome to a large extent the two main mechanisms of resistance to paclitaxel, viz. MDR resistance due to P-glycoprotein overexpression (exemplified by HCT116/VM46) and β-tubulin mutation (exemplified by A2780Tax). The test compound and paclitaxel were similarly potent in killing clonogenic cells in the two sensitive tumor cell lines (HCT116 and A2780). However, as shown in FIG. 2, against the three cell lines that had developed resistance to paclitaxel (HCT116/VM46, A2780Tax and Pat-7), the test compound performed far better than paclitaxel, almost completely retaining its cytotoxic potency against these resistant cell lines as compared to the sensitive lines.
Mechanism of Cytotoxicity—Tubulin Polymerization: The cytotoxic activities of the epothilones, like those of the taxanes, have been linked to stabilization of microtubules, which results in mitotic arrest at the G2/M transition. In this regard, the potency of the test compound was about 2.5-fold more potent than paclitaxel.
Mechanism of Cytotoxicity—Effects on Cell Cycle Progression: Similar to paclitaxel, the test compound blocks cells in the mitotic phase of the cell division cycle. Moreover, the concentration of the test compound needed to arrest cells in mitosis, as measured by flow cytometry, corresponds well to the concentration required to kill cells over the same treatment duration. Thus, as shown in FIG. 3, the test compound at a concentration close to the IC90 value (about 7.5 nM, clonogenic cytotoxicity assay) almost completely blocks cells in mitosis as early as 8 hours following the initiation of drug exposure.
Antitumor Activity by Parenteral Administration: The test compound was evaluated in a panel of eight human and murine tumor models, some of which were chosen because of their known, well-characterized resistance to paclitaxel. The tumor model characteristics are shown in Table 1 below. In addition, three paclitaxel-sensitive models were included in order to gain a full assessment of the spectrum of antitumor activity of the test compound.
TABLE 1
Paclitaxel Resistance
Tumor Histology Source Sensitivity Mechanism(s)
Human
Pat-26 Pancreatic Biopsy Insensitive Unknown
Pat-7 Ovarian Biopsy Resistant1 MDR2, MRP3
A2780Tax Ovarian Cell line Resistant Tubulin
mutation
HCT116/VM46 Colon Cell line Resistant MDR
Pat-21 Breast Biopsy Resistant1 Unknown
A2780 Ovarian Cell line Sensitive NA
HCT116 Colon Cell line Sensitive NA
LS174T Colon Cell line Sensitive NA
Murine
M5076 Fibrosarcoma Cell line Insensitive Unknown,
Non-MDR
1Clinical resistance to Taxol ®
2MDR = multidrug resistance due to P-glycoprotein overexpression
3MRP = multidrug resistance related protein
Antitumor Activity by Oral Route of Administration: Since the test compound is more stable at neutral pH than at low pH, the evaluation thereof by oral administration (PO) utilized a pH-buffering vehicle (0.25M potassium phosphate, pH 8.0). Using a every 2 days×5 schedule, the test compound was highly active orally against the Pat-7 human ovarian carcinoma model. In two separate experiments, orally administered test compound yielded 3.1 and 2.5 LCKs at its MTD. In comparison, concomitantly tested IV paclitaxel produced 1.3 and 1.2 LCK, respectively, at its optimal dose and schedule. Paclitaxel is typically inactive when administered by the oral route.
From the foregoing in vitro experimental evidence, it can be seen that the test compound retains its antineoplastic activity in cancer cells that have developed resistance to paclitaxel, whether through overexpression of the MDR P-glycoprotein or tubulin mutation. From the in vivo evidence, the test compound has clearly demonstrated antitumor activity superior to paclitaxel in both paclitaxel-resistant and sensitive tumors, and the murine fibrosarcoma M5076. The test compound was more efficacious than paclitaxel in all five paclitaxel-resistant tumors evaluated in this study (four human and one murine); viz. the clinically-derived paclitaxel resistant Pat-7 ovarian carcinoma; the A2780Tax ovarian carcinoma that is resistant to paclitaxel because of tubulin mutations; the HCT116/VM46 multidrug resistant (MDR) colon carcinoma, the clinically-derived paclitaxel-resistant Pat-21 breast carcinoma; and the murine fibrosarcoma M5076. Against three paclitaxel-sensitive human tumor xenografts, viz. A2780 human ovarians carcinoma; HCT116 and LS 174T human colon carcinoma, the test compound produced antitumor activity equivalent to paclitaxel.
A further advantage of the test compound over the prototypical taxanes is its efficacy by oral administration, producing antitumor activity when given orally that is equivalent to that produced by IV drug administration in two different human tumor xenografts.

Claims (28)

1. A method for treating a tumor in a mammal, said tumor having demonstrated resistance to oncology therapy, comprising administering to said mammal an effective amount of a composition comprising a pharmaceutically acceptable carrier and an epothilone compound of formula:
Figure USRE041393-20100622-C00009
wherein:
Q is selected from the group consisting of
Figure USRE041393-20100622-C00010
G is selected from the group consisting of alkyl, substituted alkyl, aryl, substituted aryl, heterocyclo,
Figure USRE041393-20100622-C00011
W is O or NR15;
X is O or H, H;
Y is selected from the group consisting of O; H, OR16; OR17, OR17; NOR18; H, NHOR19; H, NR20R21; H, H; and CHR22; wherein OR17, OR17 can be a cyclic ketal;
each Z1 and Z2 is, independently, selected from the group consisting of CH2, O, NR23, S, and SO2, wherein only one of Z1 and Z2 can be a heteroatom;
each B1 and B2 is, independently, selected from the group consisting of OR24, OCOR25, and O—C(═O)— NR26R27, and when B1 is H and Y is OH, H, they can form a six-membered ring ketal or acetal;
D is selected from the group consisting of NR28R29, NR30COR31 and saturated heterocycle;
each R1, R2, R3, R4, R5, R6, R7, R13, R14, R18, R19, R20, R21, R22, R26 and R27 is, independently, selected from the group consisting of H, alkyl, substituted alkyl, and aryl, and when R1 and R2 are alkyl can be joined to form a cycloalkyl, and when R3 and R4 are alkyl can be joined to form a cycloalkyl;
each R9, R10, R16, R17, R24, R25 and R31 is, independently, selected from the group consisting of H, alkyl, and substituted alkyl;
each R8, R11, R12, R28, R30, R32, and R33 is, independently, selected from the group consisting of H, alkyl, substituted alkyl, aryl, substituted aryl, cycloalkyl and heterocyclo; and
each R15, R23 and R29 is, independently, selected from the group consisting of H, alkyl, substituted alkyl, aryl, substituted aryl, cycloalkyl, heterocyclo, R32C═O, R33SO2, hydroxy, O-alkyl or O-substituted alkyl;
and pharmaceutically acceptable salts thereof and any hydrates, solvates or geometric, optical and stereoisomers thereof,
with the proviso that compounds wherein W and X are both O; R1, R2 and R7 are H; R3 R4 and R6 are methyl; R8 is H or methyl; Z1 and Z2 are CH2; G is 1-methyl-2-(substituted-4-thiazolyl)ethenyl; and Q is as defined above, are excluded.
2. The method of claim 1 wherein Q is
Figure USRE041393-20100622-C00012
X is O; Y is O; each Z1 and Z2 is, independently, CH2; and
W is NR15.
3. The method of claim 1 wherein said epothilone compound is selected from the group consisting of
[1S-[1R*,3R*(E), 7R*,10S*,11R*,12R*,16S*]]-7,11-dihydroxy-8,8,10,12,16-pentamethyl-3-[1-methyl-2-(2-methyl-4-thiazolyl)ethenyl]-4,13,17-trioxabicyclo[14.1.0]heptadecane-5,9-dione;
[1S-[1R*,3R*(E),7R*,10S*,11R*,12R*,16S*]]-7,11-dihydroxy-8,8,10,12-tetrametlnrl-3-[1-methyl-2-(2-methyl-4-thiazolyl)ethenyl]-4,13,17-trioxabicyclo[14.1.0]heptadecane-5,9-dione;
[4S-[4R*,7S*,8R*,9R*,15R*(E)]]-4,8-dihydroxy-5,5,7,9,13-pentamethyl-16-[1-methyl-2-(2-methyl-4-thiazolyl)ethenyl]-1,10-dioxa-13-cyclohexadecene-2,6-dione;
[4S-[4R*,7S*,8R*,9R*,15R*(E)]]-4,8-dihydroxy-5,5,7,9-tetramethyl-16-[1-methyl-2-(2-methyl-4-thiazolyl)ethenyl]-1,10-dioxa-13-cyclohexadecene-2,6-dione;
[1S-[1R*,3R*(E),7R*,10S*,11R*,12R*,16S*]]-7,11-dihydroxy-8,8,10,12,16-pentamethyl-3-[1-methyl-2-(2-methyl-4-thiazolyl)ethenyl]-4,14,17-trioxabicyclo[14.1.0]heptadecane-5,9-dione;
[1S-[1R*,3R*(E),7R*,10S*,11R*,12R*,16S*]]-7,11-dihydroxy-8,8,10,12-tetramethyl-3-[1-methyl-2-(2-methyl-4-thiazolyl)ethenyl]-4,14,17-trioxabicyclo[14.1.0]heptadecane-5,9-dione;
[4S-[4R*,7S*,8R*,9R*,15R*(E)]]-4,8-dihydroxy-5,5,7,9,13-pentamethyl-16-[1-methyl-2-(2-methyl-4-thiazolyl)ethenyl]-1,11-dioxa-13-cyclohexadecene-2,6-dione;
[4S-[4R*,7S*,8R*,9R*,15R*(E)]]-4,8-dihydroxy-5,5,7,9-tetramethyl-16-[1-methyl-2-(2-methyl-4-thiazolyl)ethenyl]-1,11-dioxa-13-cyclohexadecene-2,6-dione;
[1S-[1R*,3R*(E),7R*,10S*,11R*,12R*,16S*]]-7,11-dihydroxy-8,8,10,12,16-pentamethyl-3-[1-methyl-2-(2-methyl-4-thiazolyl)ethenyl]-4,17-dioxabicyclo[14.1.0]heptadecane-9-one;
[1S-[1R*,3R*(E),7R*,10S*,11R*,12R*,16S*]]-7,11-dihydroxy-8,8,10,12-tetramethyl-3-[1-methyl-2-(2-methyl-4-thiazolyl)ethenyl]-4,17-dioxabicyclo[14.1.0]heptadecane-9-one;
[1S-[1R*,3R*(E),7R*,10S*,11R*,12R*,16S*]]-7,11-dihydroxy-3,8,8,10,12,16-hexamethyl-3-[1-methyl-2-(2-methyl-4-thiazolyl)ethenyl]-4,17-dioxabicyclo[14.1.0]heptadecane-5,9-dione;
[1S-[1R*,3R*(E),7R*,10S*,11R*,12R*,16S*]]-7,11-dihydroxy-3,8,8,10,12-pentamethyl-3-[1-methyl-2-(2-methyl-4-thiazolyl)ethenyl]-4,17-dioxabicyclo[14.1.0]heptadecane-5,9-dione;
[4S-[4R*,7S*,8R*,9R*,15R*(E)]]-4,8-dihydroxy-5,5,7,9,13,16-hexamethyl-16-[1-methyl-2-(2-methyl-4-thiazolyl)ethenyl]-1-oxa-13-cyclohexadecene-2,6-dione;
[4S-[4R*,7S*,8R*,9R*,15R*(E)]]-4,8-dihydroxy-5,5,7,9,16-pentamethyl-16-[1-methyl-2-(2-methyl-4-thiazolyl)ethenyl]-1-oxa-13-cyclohexadecene-2,6-dione;
[1S-[1R*,3R*(E),7R*,10S*,11R*,12R*,16S*]]-7,11-dihydroxy-8,8,10,12,16-pentamethyl-3-[1-methyl-2-(2-methyl-4-thiazolyl)ethenyl]-4,17-dioxabicyclo[14.1.0]heptadecane-5,9-dione;
[1S-[1R*,3R*(E),7R*,10S*,11R*,12R*,16S*]]-7,11-dihydroxy-6,8,8,10,12-pentamethyl-3-[1-methyl-2-(2-methyl-4-thiazolyl)ethenyl]-4,17-dioxabicyclo[14.1.0]heptadecane-5,9-dione;
[1S-[1R*,3R*(E),7R*,10S*,11R*,12R*,16S*]]-7,11-dihydroxy-8,8,10,12,16-pentamethyl-3-[1-methyl-2-(2-methyl-4-thiazolyl)ethenyl]-4-aza-17-oxabicycllo[14.1.0]heptadecane-5,9-dione;
[1S-[1R*,3R*(E),7R*,10S*,11R*,12R*,16S*]]-7,11-dihydroxy-8,8,10,12-tetramethyl-3-[1-methyl-2-(2-methyl-4-thiazolyl)ethenyl]-4-aza-17oxabicyclo[14.1.0]heptadecane-5,9-dione;
[4S-[4R*,7S*,8R*,9R*,15R*(E)]]-4,8-dihydroxy-5,5,7,9,13-pentamethyl-16-[1-methyl-2-(2-methyl-4-thiazolyl)ethenyl]-1-aza-13-cyclohexadecene-2,6-dione;
[4S-[4R*,7S*,8R*,9R*,15R*(E)]]-4,8-dihydroxy-5,5,7,9-tetramethyl-16-[1-methyl-2-(2-methyl-4-thiazolyl)ethenyl]-1-aza-13-cyclohexadecene-2,6-dione;
[1S-1R*,3R*(E),7R*,10S*,11R*,12R*,16S*]]-7,11-dihydroxy-4,8,8,10,12,16-hexamethyl-3-[1-methyl-2-(2-methyl-4-thiazolyl)ethenyl]-4-aza-17-oxabicyclo[14.1.0]heptadecane-5,9-dione;
[1S-[1R*,3R*(E),7R*,10S*,11R*,12R*,16S*]]-7,11-dihydroxy-4,8,8,10,12-pentamethyl-3-[1-methyl-2-(2-methyl-4-thiazolyl)ethenyl]-4-aza-17-oxabicyclo[14.1.0]heptadecane-5,9-dione;
[4S-[4R*,7S*,8R*,9R*,15R*(E)]]-4,8-dihydroxy-1,5,5,7,9,13-hexamethyl-16-[1-methyl-2-(2-methyl-4-thiazolyl)ethenyl]-1-aza-13-cyclohexadecene-2,6-dione;
[4S-[4R*,7S*,8R*,9R*,15R*(E)]]-4,8-dihydroxy-1,5,5,7,9-pentamethyl-16-[1-methyl-2-(2-methyl-4-thiazolyl)ethenyl]-1-aza-13-cyclohexadecene-2,6-dione;
[1S-[1R*,3R*(E),7R*,10S*,11R*,12R*,16S*]]-7,11-dihdyroxy-8,8,10,12,16-pentamethyl-3-[1-methyl-2-(2-methyl-4-thiazolyl)ethenyl]-13-aza-4,17dioxabicyclo[14.1.0]heptadecane-5,9-dione;
[1S-[1R*,3R*(E),7R*,10S*,11R*,12R*,16S*]]-7,11-dihydroxy-8,8,10,12-tetramethyl-3-[1-methyl-2-(2-methyl-4-thiazolyl)ethenyl]-13-aza-4,17dioxabicyclo[14.1.0]heptadecane-5,9-dione;
[4S-[4R*,7S*,8R*,9R*,15R*(E)]]-4,8-dihydroxy-5,5,7,9,13-pentamethyl-16-[1-methyl-2-(2-methyl-4-thiazolyl)ethenyl]-10-aza-1-oxa-13-cyclohexadecene-2,6-dione;
[4S-[4R*,7S*,8R*,9R*,15R*(E)]]-4,8-dihydroxy-5,5,7,9-tetramethyl-16-[1-methyl-2-(2-methyl-4-thiazolyl)ethenyl]-10-aza-1-oxa-13-cyclohexadecene-2,6-dione;
[1S-[1R*,3R*(E),7R*,10S*,11R*,12R*,16S*]]-7,11-dihydroxy-8,8,10,12,16-pentamethyl-3-[1-methyl-2-(2-methyl-4-thiazolyl)ethenyl]-14-aza-4,17-dioxabicyclo[14.1.0]heptadecane-5,9-dione;
[1S-[1R*,3R*(E),7R*,10S*,11R*,12R*,16S*]]-7,11-dihydroxy-8,8,10,12-tetramethyl-3-[1-methyl-2-(2-methyl-4-thiazolyl)ethenyl]-14-aza-4,17-dioxabicyclo[14.1.0]heptanedecane-5,9-dione;
[4S-[4R*,7S*,8R*,9R*,15R*(E)]]-4,8-dihydroxy-5,5,7,9,13-pentamethyl-16-[1-methyl-2-(2-methyl-4-thiazolyl)ethenyl]-11-aza-1-oxa-13-cyclohexadecene-2,6dione;
[4S-[4R*,7S*,8R*,9R*,15R*(E)]]-4,8-dihydroxy-5,5,7,9-tetramethyl-16-[1-methyl-2-(2-methyl-4-thiazolyl)ethenyl]-11-aza-1-oxa-13-cyclohexadecene-2,6-dione;
[1S-[1R*,3R*,7R*,10S*,11R*,12R*,16S*]]-N-phenyl-7,11-dihydroxy-8,8,10,12,16-pentamethyl-5,9-dioxo-4,17-dioxabicyclo[14.1.0]heptadecane-3-carboxamide;
[1S-[1R*,3R*,7R*,10S*,11R*,12R*,16S*]]-N-phenyl-7,11-dihydroxy-8,8,10,12-tetramethyl-5,9-dioxo-4,17-dioxabicyclo[14.1.0]heptadecane-3-carboxamide;
[4S-[4R*,7S*,8R*,9R*,15R*]]-N-phenyl-4,8-dihydroxy-5,5,7,9,13-pentamethyl-2,6-dioxo-1-oxa-13-cyclohexadecene-16-carboxamide;
[4S-[4R*,7S*,8R*,9R*,15R*]]-N-phenyl-4,8-dihydroxy-5,5,7,9-tetramethyl-2,6-dioxo-1-oxa-13-cyclohexadecene-16-carboxamide;
[1S-[1R*,3R*(E),7R*,10S*,11R*,12R*,16S*]]-7,11-dihydroxy-8,8,10,12,16-pentamethyl-3-[1-methyl-2-(2-methyl-4-thiazolyl)cyclopropyl]-4,17-dioxabicyclo[14.1.0]heptadecane-5,9-dione;
[1S-[1R*,3R*(E),7R*,10S*,11R*,12R*,16S*]]-7,11-dihydroxy-8,8,10,12-tetramethyl-3-[1-methyl-2-(2-methyl-4-thiazolyl)cyclopropyl]-4,17-dioxabicyclo[14.1.0]heptadecane-5,9-dione; and
[4S-[4R*,7S*,8R*,9R*,15R*(E)]]-4,8-dihydroxy-5,5,7,9,13-pentamethyl-16-[1-methyl-2-(2-hydroxymethyl-4-thiazolyl)ethenyl]-1-aza-13(Z)-cyclohexadecene-2,6-dione;
and pharmaceutically acceptable salts, solvates and hydrates thereof.
4. The method of claim 1 wherein said epothilone compound is of formula:
Figure USRE041393-20100622-C00013
5. The method of claim 1 wherein said mammal is a human.
6. The method of claim 1 wherein the composition containing said epothilone compound is administered parenterally.
7. The method of claim 6 wherein said epothilone compound is of formula:
Figure USRE041393-20100622-C00014
8. The method of claim 1 wherein the composition containing said epothilone compound is administered orally.
9. The method of claim 8 wherein said epothilone compound is of formula:
Figure USRE041393-20100622-C00015
10. The method of claim 1 wherein said tumor was initially not responsive to oncology therapy.
11. The method of claim 1 wherein said tumor was initially responsive to oncology therapy, but developed resistance thereto during the course of treatment.
12. The method of claim 1 wherein said compound is administered simultaneously or sequentially with a chemotherapeutic agent useful in the treatment of cancer or other proliferative diseases.
13. The method of claim 1 wherein the oncology therapy is a taxane.
14. The method of claim 1 wherein the oncology therapy is paclitaxel.
15. The method of claim 1 wherein the tumor is of the bladder, breast, colon, kidney, liver, lung, ovary, pancreas, stomach, cervix, thyroid or skin.
16. A method for treating a tumor in a mammal, said tumor being resistant to oncology therapy with a taxane, comprising administering to said mammal an effective amount of a composition comprising a pharmaceutically acceptable carrier and a compound having the formula,
Figure USRE041393-20100622-C00016
17. The method of claim 16 wherein said mammal is a human.
18. The method of claim 17 wherein said tumor was initially not responsive to taxane therapy.
19. The method of claim 17, wherein said tumor was initially responsive to taxane therapy, but developed resistance thereto during the course of treatment.
20. The method of claim 17, wherein said tumor is innately resistant to taxane therapy.
21. The method of claim 17, wherein the taxane is paclitaxel.
22. The method of claim 17 wherein the tumor is of the bladder, breast, colon, kidney, liver, lung, ovary, pancreas, stomach, cervix, thyroid or skin.
23. The method of claim 22, wherein the tumor is of the breast.
24. The method of claim 22, wherein the tumor is of the pancreas.
25. The method of claim 23, wherein the oncology therapy is paclitaxel.
26. The method of claim 23 wherein said tumor was initially not responsive to taxane therapy.
27. The method of claim 23 wherein said tumor was initially responsive to taxane therapy, but developed resistance thereto during the course of treatment.
28. The method of claim 23, wherein said tumor is innately resistant to taxane therapy.
US11/346,579 2001-02-20 2006-02-02 Treatment of refractory tumors using epothilone derivatives Expired - Lifetime USRE41393E1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US11/346,579 USRE41393E1 (en) 2001-02-20 2006-02-02 Treatment of refractory tumors using epothilone derivatives

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US26983601P 2001-02-20 2001-02-20
US10/072,123 US6686380B2 (en) 2001-02-20 2002-02-08 Treatment of refractory tumors using epothilone derivatives
US11/346,579 USRE41393E1 (en) 2001-02-20 2006-02-02 Treatment of refractory tumors using epothilone derivatives

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US10/072,123 Reissue US6686380B2 (en) 2001-02-20 2002-02-08 Treatment of refractory tumors using epothilone derivatives

Publications (1)

Publication Number Publication Date
USRE41393E1 true USRE41393E1 (en) 2010-06-22

Family

ID=23028846

Family Applications (2)

Application Number Title Priority Date Filing Date
US10/072,123 Ceased US6686380B2 (en) 2001-02-20 2002-02-08 Treatment of refractory tumors using epothilone derivatives
US11/346,579 Expired - Lifetime USRE41393E1 (en) 2001-02-20 2006-02-02 Treatment of refractory tumors using epothilone derivatives

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US10/072,123 Ceased US6686380B2 (en) 2001-02-20 2002-02-08 Treatment of refractory tumors using epothilone derivatives

Country Status (18)

Country Link
US (2) US6686380B2 (en)
EP (1) EP1385529A4 (en)
JP (1) JP2004522774A (en)
KR (1) KR20040025895A (en)
CN (1) CN1774253A (en)
BG (1) BG108075A (en)
BR (1) BR0207487A (en)
CA (1) CA2438610A1 (en)
EE (1) EE200300396A (en)
HU (1) HUP0303175A2 (en)
IL (1) IL156988A0 (en)
IS (1) IS6918A (en)
MX (1) MXPA03007394A (en)
NO (1) NO20033684L (en)
PL (1) PL363362A1 (en)
RU (1) RU2003128311A (en)
WO (1) WO2002066038A1 (en)
ZA (1) ZA200306173B (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2021142202A1 (en) 2020-01-10 2021-07-15 R-Pharm Us Operating Llc Compositions of ixabepilone

Families Citing this family (25)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2273083C (en) 1996-12-03 2012-09-18 Sloan-Kettering Institute For Cancer Research Synthesis of epothilones, intermediates thereto, analogues and uses thereof
US20050043376A1 (en) * 1996-12-03 2005-02-24 Danishefsky Samuel J. Synthesis of epothilones, intermediates thereto, analogues and uses thereof
US6204388B1 (en) * 1996-12-03 2001-03-20 Sloan-Kettering Institute For Cancer Research Synthesis of epothilones, intermediates thereto and analogues thereof
US6605599B1 (en) 1997-07-08 2003-08-12 Bristol-Myers Squibb Company Epothilone derivatives
US6780620B1 (en) * 1998-12-23 2004-08-24 Bristol-Myers Squibb Company Microbial transformation method for the preparation of an epothilone
US20020058286A1 (en) * 1999-02-24 2002-05-16 Danishefsky Samuel J. Synthesis of epothilones, intermediates thereto and analogues thereof
CZ20032022A3 (en) 2001-01-25 2004-04-14 Bristol-Myers Squibb Company Parenteral formulation containing epothilone analogs
SK11082003A3 (en) * 2001-03-14 2004-08-03 Bristol-Myers Squibb Company Combination of epotilone analogues and chemotherapeutic agents for treatment of proliferative diseases
TW200403994A (en) * 2002-04-04 2004-03-16 Bristol Myers Squibb Co Oral administration of EPOTHILONES
TW200400191A (en) * 2002-05-15 2004-01-01 Bristol Myers Squibb Co Pharmaceutical compositions and methods of using C-21 modified epothilone derivatives
US6921769B2 (en) 2002-08-23 2005-07-26 Sloan-Kettering Institute For Cancer Research Synthesis of epothilones, intermediates thereto and analogues thereof
CA2496477C (en) * 2002-08-23 2012-10-16 Sloan-Kettering Institute For Cancer Research Synthesis of epothilones, intermediates thereto, analogues and uses thereof
US7649006B2 (en) 2002-08-23 2010-01-19 Sloan-Kettering Institute For Cancer Research Synthesis of epothilones, intermediates thereto and analogues thereof
WO2004032866A2 (en) 2002-10-09 2004-04-22 Kosan Biosciences, Inc. Therapeutic formulations
TW200503738A (en) * 2003-07-16 2005-02-01 Tzu Chi Buddhist General Hospital Method for extracting antineoplastic components from bupleurum scorzonerifolium
US20050171167A1 (en) * 2003-11-04 2005-08-04 Haby Thomas A. Process and formulation containing epothilones and analogs thereof
US20090004277A1 (en) * 2004-05-18 2009-01-01 Franchini Miriam K Nanoparticle dispersion containing lactam compound
WO2006017761A2 (en) * 2004-08-05 2006-02-16 Emory University Epothilone analogues as therapeutic agents
WO2006055742A1 (en) * 2004-11-18 2006-05-26 Bristol-Myers Squibb Company Enteric coated bead comprising epothilone or epothilone analog, and preparation and administration thereof
KR20070084325A (en) 2004-11-18 2007-08-24 브리스톨-마이어스 스큅 컴퍼니 Enteric coated bead comprising ixabepilone, and preparation thereof
AU2006336468B2 (en) 2005-02-11 2012-04-12 University Of Southern California Method of expressing proteins with disulfide bridges
EP2029156A4 (en) * 2006-05-01 2010-07-21 Univ Southern California Combination therapy for treatment of cancer
US8463852B2 (en) * 2006-10-06 2013-06-11 Oracle International Corporation Groupware portlets for integrating a portal with groupware systems
US8802394B2 (en) 2008-11-13 2014-08-12 Radu O. Minea Method of expressing proteins with disulfide bridges with enhanced yields and activity
EP2571525A4 (en) 2010-05-18 2016-04-27 Cerulean Pharma Inc Compositions and methods for treatment of autoimmune and other diseases

Citations (53)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1993010121A1 (en) 1991-11-19 1993-05-27 GESELLSCHAFT FüR BIOTECHNOLOGISCHE FORSCHUNG MBH (GBF) Epothilones, process for preparing the same and their use as medicaments and as plant protecting agents
DE19542986A1 (en) 1995-11-17 1997-05-22 Biotechnolog Forschung Gmbh New epothilone derivatives useful as cytostatics
WO1997019086A1 (en) 1995-11-17 1997-05-29 GESELLSCHAFT FüR BIOTECHNOLOGISCHE FORSCHUNG MBH (GBF) Epothilone derivatives, preparation and use
DE19636343C1 (en) 1996-08-30 1997-10-23 Schering Ag New (di:methyl)-dioxanyl-methyl-pentanone and related compounds
WO1998008849A1 (en) 1996-08-30 1998-03-05 Novartis Aktiengesellschaft Method for producing epothilones, and intermediate products obtained during the production process
DE19639456A1 (en) 1996-09-25 1998-03-26 Biotechnolog Forschung Gmbh New epothilone derivatives
DE19645362A1 (en) 1996-10-28 1998-04-30 Ciba Geigy Ag Production of epothilone compounds with taxol-like activity
DE19645361A1 (en) 1996-08-30 1998-04-30 Ciba Geigy Ag Production of epothilone compounds with taxol-like activity
WO1998022461A1 (en) 1996-11-18 1998-05-28 GESELLSCHAFT FüR BIOTECHNOLOGISCHE FORSCHUNG MBH (GBF) Epothilone c, d, e and f, production process, and their use as cytostatic as well as phytosanitary agents
WO1998024427A2 (en) 1996-12-02 1998-06-11 Angiotech Pharmaceuticals, Inc. Compositions and methods for treating or preventing inflammatory diseases
WO1998025929A1 (en) 1996-12-13 1998-06-18 Novartis Ag Epothilone analogs
DE19701758A1 (en) 1997-01-20 1998-07-23 Wessjohann Ludgar A Dr New beta-keto-alcohol derivatives
WO1998038192A1 (en) 1997-02-25 1998-09-03 GESELLSCHAFT FüR BIOTECHNOLOGISCHE FORSCHUNG MBH (GBF) Epothilones with a modified side chain
WO1998039192A1 (en) 1997-03-06 1998-09-11 Harry Nijenhuis Management B.V. Terminal for transferring containers, and a container car
DE19713970A1 (en) 1997-04-04 1998-10-08 Ludger A Dr Wessjohann New structural elements which include prenyl derivatives
WO1998047891A1 (en) 1997-04-18 1998-10-29 Studiengesellschaft Kohle Mbh Selective olefin metathesis of bifunctional or polyfunctional substrates in compressed carbon dioxide as reaction medium
DE19720312A1 (en) 1997-05-15 1998-11-19 Hoechst Ag Preparation with increased in vivo tolerance
DE19821954A1 (en) 1997-05-15 1998-11-19 Biotechnolog Forschung Gmbh Preparation of epothilone derivatives
DE19726627A1 (en) 1997-06-17 1998-12-24 Schering Ag New intermediates for epothilone
WO1999001124A1 (en) 1996-12-03 1999-01-14 Sloan-Kettering Institute For Cancer Research Synthesis of epothilones, intermediates thereto, analogues and uses thereof
WO1999002514A2 (en) 1997-07-08 1999-01-21 Bristol-Myers Squibb Company Epothilone derivatives
WO1999003848A1 (en) 1997-07-16 1999-01-28 Schering Aktiengesellschaft Thiazole derivatives, method for their production and use
WO1999007692A2 (en) 1997-08-09 1999-02-18 Schering Aktiengesellschaft New epothilone derivatives, method for producing same and their pharmaceutical use
WO1999027890A2 (en) 1997-12-04 1999-06-10 Bristol-Myers Squibb Company A process for the preparation of ring-opened epothilone intermediates which are useful for the preparation of epothilone analogs
WO1999039694A2 (en) 1998-02-05 1999-08-12 Novartis Ag Compositions containing organic compounds
WO1999042602A2 (en) 1998-02-19 1999-08-26 Novartis Ag Fermentative preparation process for cytostatics and crystal forms thereof
WO1999043320A1 (en) 1998-02-25 1999-09-02 Novartis Ag Use of epothilones for the treatment of cancer
WO1999043653A1 (en) 1998-02-25 1999-09-02 Sloan-Kettering Institute For Cancer Research Synthesis of epothilones, intermediates thereto and analogues therof
US5969145A (en) 1996-08-30 1999-10-19 Novartis Ag Process for the production of epothilones and intermediate products within the process
WO1999054319A1 (en) 1998-04-21 1999-10-28 Bristol-Myers Squibb Company 12,13-modified epothilone derivatives
WO1999067252A2 (en) 1998-06-22 1999-12-29 Novartis Ag Epothilone derivatives and their synthesis and use
WO2000000485A1 (en) 1998-06-30 2000-01-06 Schering Aktiengesellschaft Epothilon derivatives, their preparation process, intermediate products and their pharmaceutical use
WO2000031247A2 (en) 1998-11-20 2000-06-02 Kosan Biosciences, Inc. Recombinant methods and materials for producing epothilone and epothilone derivatives
WO2000037473A1 (en) 1998-12-22 2000-06-29 Novartis Ag Epothilone derivatives and their use as antitumor agents
WO2000049021A2 (en) 1999-02-18 2000-08-24 Schering Aktiengesellschaft 16-halogen-epothilone derivatives, method for producing them and their pharmaceutical use
WO2000050423A1 (en) 1999-02-22 2000-08-31 Gesellschaft Fuer Biotechnologische Forschung Mbh (Gbf) C-21 modified epothilones
WO2000068589A1 (en) 1999-05-06 2000-11-16 Transrol Torque-transmitting ball joint
US6204388B1 (en) 1996-12-03 2001-03-20 Sloan-Kettering Institute For Cancer Research Synthesis of epothilones, intermediates thereto and analogues thereof
US6211412B1 (en) 1999-03-29 2001-04-03 The University Of Kansas Synthesis of epothilones
US6302838B1 (en) 1998-02-25 2001-10-16 Novartis Ag Cancer treatment with epothilones
US20020010213A1 (en) * 1994-08-31 2002-01-24 Sawyer Jason Scott Methods for identifying and treating resistant tumors
US20020143038A1 (en) 2001-01-25 2002-10-03 Rebanta Bandyopadhyay Parenteral formulation for epothilone analogs
US20020160354A1 (en) * 1997-12-01 2002-10-31 Myles C. Cabot Methods for screening therapeutically effective agents
US20020177615A1 (en) 2001-01-25 2002-11-28 Rebanta Bandyopadhyay Pharmaceutical compositions, dosage forms and methods for oral administration of epothilones
US6518421B1 (en) 2000-03-20 2003-02-11 Bristol-Myers Squibb Company Process for the preparation of epothilone analogs
US20030073677A1 (en) 2001-03-14 2003-04-17 Lee Francis Y.F. Combination of epothilone analogs and chemotherapeutic agents for the treatment of proliferative diseases
US6593115B2 (en) 2000-03-24 2003-07-15 Bristol-Myers Squibb Co. Preparation of epothilone intermediates
US6670384B2 (en) 2001-01-25 2003-12-30 Bristol-Myers Squibb Company Methods of administering epothilone analogs for the treatment of cancer
US20040024032A1 (en) 2002-06-14 2004-02-05 Maurizio Voi Combination of epothilone analogs and chemotherapeutic agents for the treatment of proliferative diseases
US6689802B2 (en) 2000-08-16 2004-02-10 Bristol-Myers Squibb Company Polymorphs of an epothilone analog
US20040076177A1 (en) 2002-10-21 2004-04-22 Zeke Koch Method and system to shut down and control computer radios
US20040132146A1 (en) 2002-09-23 2004-07-08 Daniel Benigni Methods for the preparation, isolation and purification of epothilone B, and x-ray crystal structures of epothilone B
US6867305B2 (en) 1996-12-03 2005-03-15 Sloan-Kettering Institute For Cancer Research Synthesis of epothilones, intermediates thereto and analogues thereof

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
PE20010116A1 (en) 1999-04-30 2001-02-15 Schering Ag 6-ALKENYL-, 6-ALKINYL- AND 6-EPOXY-EPOTILONE DERIVATIVES, PROCEDURES FOR THEIR PREPARATION

Patent Citations (67)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1993010121A1 (en) 1991-11-19 1993-05-27 GESELLSCHAFT FüR BIOTECHNOLOGISCHE FORSCHUNG MBH (GBF) Epothilones, process for preparing the same and their use as medicaments and as plant protecting agents
DE4138042A1 (en) 1991-11-19 1993-05-27 Biotechnolog Forschung Gmbh EPOTHILONES, THEIR PRODUCTION PROCESS AND MEANS CONTAINING THEM
US20020010213A1 (en) * 1994-08-31 2002-01-24 Sawyer Jason Scott Methods for identifying and treating resistant tumors
DE19542986A1 (en) 1995-11-17 1997-05-22 Biotechnolog Forschung Gmbh New epothilone derivatives useful as cytostatics
WO1997019086A1 (en) 1995-11-17 1997-05-29 GESELLSCHAFT FüR BIOTECHNOLOGISCHE FORSCHUNG MBH (GBF) Epothilone derivatives, preparation and use
DE19636343C1 (en) 1996-08-30 1997-10-23 Schering Ag New (di:methyl)-dioxanyl-methyl-pentanone and related compounds
WO1998008849A1 (en) 1996-08-30 1998-03-05 Novartis Aktiengesellschaft Method for producing epothilones, and intermediate products obtained during the production process
US5969145A (en) 1996-08-30 1999-10-19 Novartis Ag Process for the production of epothilones and intermediate products within the process
DE19645361A1 (en) 1996-08-30 1998-04-30 Ciba Geigy Ag Production of epothilone compounds with taxol-like activity
DE19639456A1 (en) 1996-09-25 1998-03-26 Biotechnolog Forschung Gmbh New epothilone derivatives
DE19645362A1 (en) 1996-10-28 1998-04-30 Ciba Geigy Ag Production of epothilone compounds with taxol-like activity
WO1998022461A1 (en) 1996-11-18 1998-05-28 GESELLSCHAFT FüR BIOTECHNOLOGISCHE FORSCHUNG MBH (GBF) Epothilone c, d, e and f, production process, and their use as cytostatic as well as phytosanitary agents
WO1998024427A2 (en) 1996-12-02 1998-06-11 Angiotech Pharmaceuticals, Inc. Compositions and methods for treating or preventing inflammatory diseases
US6316630B1 (en) 1996-12-03 2001-11-13 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
US6242469B1 (en) 1996-12-03 2001-06-05 Sloan-Kettering Institute For Cancer Research Synthesis of epothilones, intermediates thereto, 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
WO1999001124A1 (en) 1996-12-03 1999-01-14 Sloan-Kettering Institute For Cancer Research Synthesis of epothilones, intermediates thereto, analogues and uses thereof
US6369234B1 (en) 1996-12-03 2002-04-09 Sloan-Kettering Institute For Cancer Research Synthesis of epothilones, intermediates thereto, analogues and uses thereof
WO1998025929A1 (en) 1996-12-13 1998-06-18 Novartis Ag Epothilone analogs
DE19701758A1 (en) 1997-01-20 1998-07-23 Wessjohann Ludgar A Dr New beta-keto-alcohol derivatives
WO1998038192A1 (en) 1997-02-25 1998-09-03 GESELLSCHAFT FüR BIOTECHNOLOGISCHE FORSCHUNG MBH (GBF) Epothilones with a modified side chain
WO1998039192A1 (en) 1997-03-06 1998-09-11 Harry Nijenhuis Management B.V. Terminal for transferring containers, and a container car
DE19713970A1 (en) 1997-04-04 1998-10-08 Ludger A Dr Wessjohann New structural elements which include prenyl derivatives
WO1998047891A1 (en) 1997-04-18 1998-10-29 Studiengesellschaft Kohle Mbh Selective olefin metathesis of bifunctional or polyfunctional substrates in compressed carbon dioxide as reaction medium
EP0879605A2 (en) 1997-05-15 1998-11-25 Hoechst Aktiengesellschaft Glycosyl prodrug conjugate with enhanced tolerance
DE19821954A1 (en) 1997-05-15 1998-11-19 Biotechnolog Forschung Gmbh Preparation of epothilone derivatives
DE19720312A1 (en) 1997-05-15 1998-11-19 Hoechst Ag Preparation with increased in vivo tolerance
DE19726627A1 (en) 1997-06-17 1998-12-24 Schering Ag New intermediates for epothilone
US6605599B1 (en) 1997-07-08 2003-08-12 Bristol-Myers Squibb Company Epothilone derivatives
WO1999002514A2 (en) 1997-07-08 1999-01-21 Bristol-Myers Squibb Company Epothilone derivatives
WO1999003848A1 (en) 1997-07-16 1999-01-28 Schering Aktiengesellschaft Thiazole derivatives, method for their production and use
WO1999007692A2 (en) 1997-08-09 1999-02-18 Schering Aktiengesellschaft New epothilone derivatives, method for producing same and their pharmaceutical use
US20020160354A1 (en) * 1997-12-01 2002-10-31 Myles C. Cabot Methods for screening therapeutically effective agents
US6365749B1 (en) 1997-12-04 2002-04-02 Bristol-Myers Squibb Company Process for the preparation of ring-opened epothilone intermediates which are useful for the preparation of epothilone analogs
WO1999027890A2 (en) 1997-12-04 1999-06-10 Bristol-Myers Squibb Company A process for the preparation of ring-opened epothilone intermediates which are useful for the preparation of epothilone analogs
WO1999039694A2 (en) 1998-02-05 1999-08-12 Novartis Ag Compositions containing organic compounds
WO1999042602A2 (en) 1998-02-19 1999-08-26 Novartis Ag Fermentative preparation process for cytostatics and crystal forms thereof
US6194181B1 (en) 1998-02-19 2001-02-27 Novartis Ag Fermentative preparation process for and crystal forms of cytostatics
US6302838B1 (en) 1998-02-25 2001-10-16 Novartis Ag Cancer treatment with epothilones
WO1999043320A1 (en) 1998-02-25 1999-09-02 Novartis Ag Use of epothilones for the treatment of cancer
WO1999043653A1 (en) 1998-02-25 1999-09-02 Sloan-Kettering Institute For Cancer Research Synthesis of epothilones, intermediates thereto and analogues therof
WO1999054319A1 (en) 1998-04-21 1999-10-28 Bristol-Myers Squibb Company 12,13-modified epothilone derivatives
WO1999067252A2 (en) 1998-06-22 1999-12-29 Novartis Ag Epothilone derivatives and their synthesis and use
WO2000000485A1 (en) 1998-06-30 2000-01-06 Schering Aktiengesellschaft Epothilon derivatives, their preparation process, intermediate products and their pharmaceutical use
WO2000031247A2 (en) 1998-11-20 2000-06-02 Kosan Biosciences, Inc. Recombinant methods and materials for producing epothilone and epothilone derivatives
WO2000037473A1 (en) 1998-12-22 2000-06-29 Novartis Ag Epothilone derivatives and their use as antitumor agents
WO2000049021A2 (en) 1999-02-18 2000-08-24 Schering Aktiengesellschaft 16-halogen-epothilone derivatives, method for producing them and their pharmaceutical use
US6262094B1 (en) 1999-02-22 2001-07-17 Bristol-Myers Squibb Company C-21 modified epothilones
WO2000050423A1 (en) 1999-02-22 2000-08-31 Gesellschaft Fuer Biotechnologische Forschung Mbh (Gbf) C-21 modified epothilones
US6211412B1 (en) 1999-03-29 2001-04-03 The University Of Kansas Synthesis of epothilones
WO2000068589A1 (en) 1999-05-06 2000-11-16 Transrol Torque-transmitting ball joint
US6518421B1 (en) 2000-03-20 2003-02-11 Bristol-Myers Squibb Company Process for the preparation of epothilone analogs
US6593115B2 (en) 2000-03-24 2003-07-15 Bristol-Myers Squibb Co. Preparation of epothilone intermediates
US20040023345A1 (en) 2000-03-24 2004-02-05 Vite Gregory D. Preparation of epothilone intermediates
US6982276B2 (en) 2000-08-16 2006-01-03 Bristol-Myers Squibb Company Polymorphs of an epothilone analog
US6689802B2 (en) 2000-08-16 2004-02-10 Bristol-Myers Squibb Company Polymorphs of an epothilone analog
US20020177615A1 (en) 2001-01-25 2002-11-28 Rebanta Bandyopadhyay Pharmaceutical compositions, dosage forms and methods for oral administration of epothilones
US6670384B2 (en) 2001-01-25 2003-12-30 Bristol-Myers Squibb Company Methods of administering epothilone analogs for the treatment of cancer
US6576651B2 (en) 2001-01-25 2003-06-10 Bristol-Myers Squibb Company Pharmaceutical compositions, dosage forms and methods for oral administration of epothilones
US20020143038A1 (en) 2001-01-25 2002-10-03 Rebanta Bandyopadhyay Parenteral formulation for epothilone analogs
US20040214871A1 (en) 2001-03-14 2004-10-28 Lee Francis Y F Combination of epothilone analogs and chemotherapeutic agents for the treatment of proliferative diseases
US20030073677A1 (en) 2001-03-14 2003-04-17 Lee Francis Y.F. Combination of epothilone analogs and chemotherapeutic agents for the treatment of proliferative diseases
US20050159461A1 (en) 2001-03-14 2005-07-21 Lee Francis Y. Combination of epothilone analogs and chemotherapeutic agents for the treatment of prolferative diseases
US20040024032A1 (en) 2002-06-14 2004-02-05 Maurizio Voi Combination of epothilone analogs and chemotherapeutic agents for the treatment of proliferative diseases
US20040132146A1 (en) 2002-09-23 2004-07-08 Daniel Benigni Methods for the preparation, isolation and purification of epothilone B, and x-ray crystal structures of epothilone B
US20040076177A1 (en) 2002-10-21 2004-04-22 Zeke Koch Method and system to shut down and control computer radios

Non-Patent Citations (74)

* Cited by examiner, † Cited by third party
Title
Abstract and Slides: Lee, Francis, et al., "An Epothilone Analog Possessing Potent Activity Against Paclitaxel Sensitive and Resistant Human Tumors", Proceedings of the American Association for Cancer Research, 91stAnn. Meeting. San Francisco, CA, Apr. 1-5, 2000.
Altman, K. H., "Synthesis and Biological Evaluation of Highly Potent Analogues or Epothilones B and D", Bioorganic & Med. Chem. Ltrs. 10 (2000), pp. 2765-2768.
Altmann, K.H., et al., "Epothilones and Related Structures-A New Class of Microtubule Inhibitors With Patent In Vivo Antitumor Activity", Biochim. Biophys Acta, 1470 (2000).
Balog, A., et al., "Total Synthesis of (-)-Epothilone A", Angew. Chem. Int. Ed. Eng., vol. 35, No. 23/24, 2801-2803 (1996).
Bertinato, P., et al., "Studies Toward a Synthesis of Epothilone A: Stereocontrolled Assembly of the Acyl Region and Models for Macrocyclization", J. Org. Chem., vol. 61, No. 23, 8000-8001 (1996).
Bertini, F., et al., "Alkenes from Epoxides by Reductive Elimination with Magnesium Bromide-Magnesium Amalgam", Chem. Commun., 144 (1970).
Blagosklonny, M.V., Leukemia, vol. 13, 1999, pp. 2031-2035.
Bollag, D., "Epothilones: Novel Microtubule-Stabilising Agents", Expert Opin. Invest. Drugs, vol. 6, No. 7, 867-873 (1997).
Bollag, D., et al., "Epothilone, A New Structural Class of Microtubule Stabilizer", Abstract, Proc. Am. Assoc. Cancer Res., vol. 36, 86 Meet. 454 (1995).
Bollag, D.M., et al., "Epothilones, A New Class of Microtubule-stabilizing Agents with a Taxol-like Mechanism of Action", Cancer Res.55, No. 11, 2325-2333 (1995).
Borzilleri, R. M., et al., J. Am.Chem. Soc., vol. 122, 2000, pp. 8890-8897.
Chemical & Engineering News, "Epothilone Epiphany: Total Synthesis", vol. 74, No. 52, 24-26 (1996).
Chemical & Engineering News, "First Total Synthesis of Epothilone B", vol. 75, No. 13, 23 (1997).
Chemical & Engineering News, "Solid-Phase Epothilone Synthesis Used to Create Analog Library", vol. 75, No. 20, 33 (1997).
Chou et al., 1998, Desoxyepothilone B is curative against human tumor xenografts that are refractory to paclitaxel, Proc. Natl. Acad. Sci. USA 95: 15798-15802.
Chou, T., et al., "Desoxyepothilone B: An Efficacious Microtubule-targeted Antitumor Agent with a Promising In Vivo Profile Relative to Epothilone B" PNAS, vol. 95, Aug. 1998, pp. 9642-9647.
Claus, E., et al., "Synthesis of the C1-C9 Segment of Epothilons", Tetrahedron Lett., vol. 38, No. 8, 1359-1362 (1997).
Danishefsky et al. "Remote Effects in Macrolide Formation through Ring-Forming Olefin Metathesis: An Application to the Synthesis of Fully Active Epothilone Congeners", J. Am. Chem. Soc., vol. 119, (1997) pp. 2733-2734.
De Brabander, J., et al., "Towards a Synthesis of Epothilone A: Rapid Assembly of the C1-C6 and C7-C12 Fragments", Synlett, vol. 7, 824-826 (1997).
Fujisawa, T., et al., "Deoxygenation of Epoxides to Olefins with FeCl3-n-BuLi System", Chem. Lett., 883-886 (1974).
Fujiwara, Y., et al., "Reductive Coupling of Carbonyl Compounds to Olefins by Tungsten Hexachloride-Lithium Aluminum Hydride and Some Tungsten and Molybdenum Carbonyls", J. Org. Chem., vol. 43, No. 12, 2477-2479 (1978).
Gabriel, T. and Wessjohann, L., "The Chromium-Reformatsky Reaction: Asymmetric Synthesis of the Aldol Fragment of the Cytotoxic Epothilons from 3-(2-Bromoacyl)-2-Oxazolidinones", Tetrahedron Lett., vol. 38, No. 8, 1363-1366 (1997).
Gerth, et al. "Studies on the Biosynthesis of Epothilones: The Biosynthetic Origin of the Carbon Skelton", Journal of Antibiotics, vol. 53, No. 12, Dec. 2000, pp. 1373-1377.
Gerth, K., et al., "Epothilons A and B: Antifungal and Cytotoxic Compounds from Sorangiusm cellulosum(Myxobacteria) Production, Physico-chemical and Biological Properties", J. Antibiotics, vol. 49, No. 6, 560-563 (1996).
Gladysz, J.A., et al., "Deoxygenation of Epoxides by Metal Atom Cocondensation", J. Org. Chem., vol. 41, No. 22, 3647-3648 (1976).
Hardt, et al., "New Natural Epothilones from Sorangium cellulosum, Strains So ce90/B2 and So ce90/D13: Isolation, Structure Elucidation and SAR Studies", J. of Natural Products, vol. 64, No. 7, (2001) pp. 847-856.
Hoffe, G., et al., "Epothilone A and B-Novel 16-Membered Macrolides with Cytotoxic Activity: Isolation, Crystal Structure, and Conformation in Solution", Angew. Chem. Int. Ed. Engl., vol. 35, No. 13/14, 1567-1569 (1996).
Hoffe, G., et al., "N-Oxidation of Epothilone A-C and O-Acyl Rearrangement to C-19 and C-21-Substituted Epothilones", Angew. Chem. Int. Ed., vol. 38, No. 13/14, 1971-1974 (1999).
Inokuchi, T., et al., "Opening of Epoxides to Olefins or Halohydrins with Vanadium (ll)-Tetrahydrofuran or Vanadium (III)-Tetrahydrofuran Complexes", Synlett, No. 6, 510-512 (1992).
Kowalski, R. J., et al., "Activities of the Microtubule-stablizing Agents Epothilones A and B with Purified Tubulin and in Cells Resistant to Paclitaxel (Taxol®)" J. Biol. Chem., vol. 272, No. 4, 2534-2541 (1997).
Kupchan, S. M., et al., "Reductive Elimination of Epoxides to Olefins with Zinc-Coppper Couple", J. Org. Chem., vol. 36, No. 9, 1187-1190 (1971).
Lee, F.Y.F. et al., Clinical Caner Research, vol. 7, May 2001 (May 2001) pp. 1429-1437.
Marshall, A., "Total Synthesis of Epothilone", Nature Biotechnology, vol. 15, No. 3, 205 (1997).
Martin, M. G., et al., "Epoxides as Alkene Protecting Groups. A Mild and Efficient Deoxygenation", Tetrahedron Letters, vol. 25, No. 3, 251-254 (1984).
McMurry, J. E., et al., "Reduction of Epoxides to Olefins with Low Valent Titanium", J. Org. Chem., vol. 40, No. 17, 2555-2556 (1975).
McMurry, J.E., et al., "Some Deoxygenation Reactions with Low-Valent Titanium (TiCL/LiAIH4 )", J. Org. Chem., vol. 43, No. 17, 3249-3254.
Meng, D., et al., "Remote Effects in Macrolide Formation Through Ring-Forming Olefin Methathesis: An Application to the Synthesis of Fully Active Epothilone Congeners", J. Am. Chem. Soc., vol. 119, No. 11, 2733-2734 (1997).
Meng, D., et al., "Studies Toward a Syntheses of Epothilone A: Use of Hydropyran Templates for the Management of Acyclic Stereochemical Relationships", J. Org. Chem.vol. 61, No. 23, 7998-7999 (1996).
Meng, D., et al., "Total Synthesis of Epothilones A and B", J. Am. Chem. Soc., vol. 119, No. 42, 10073-10092 (1997).
Mensching, S. and Kalesse, M., "Generation of Thiazoles by Column Dehydrogenation of Thiazolidines with MnO2", J. Prakt. Chem., vol. 339, No. 1, 96-97 (1997).
Mulzer, J. and Mantoulidis, A., "Synthesis of the C(1)-C(9) Segment of the Cytotoxic Macrolides Epothilon A and B", Tetrahedron Lett., vol. 37, No. 51, 9179-9182 (1996).
Nicolaou et al., "Chemical Biology of Epothilones", Angew. Chem. Int. Ed., 37, 2014-2045 (1988).
Nicolaou et al., "Total Synthesis of Epothilone E and Analogs with Modified Side Chains Through The Stille Coupling Reaction", Angew. Chem. Int. Ed. 37, 84-87 (1998).
Nicolaou et al., "Total Synthesis of Oxazole-and Cyclopropane-Containing Epothilone B Analogues by the Macrolactorization Approach", Chemistry, European Journal, vol. 3, No. 12, 1971-1986 (1997).
Nicolaou, K. C., et al., "An Approach to Epothilones Based on Olefin Metathesis", Angew. Chem. Int. Ed. Engl., vol. 35, No. 20, 2399-2401(1996).
Nicolaou, K. C., et al., "Designed Epothilones: Combinatorial Synthesis, Tubulin Assembly Properties, and Cytotoxic Action against Taxol-Resistant Tumor Cells", Angew. Chem. Int. Ed. Engl., vol. 36, No. 19, 2097-2103 (1997).
Nicolaou, K. C., et al., "Synthesis and Biological Properties of C12, 13-Cycloprpylpeothilone A and Related Epothilones", Chemistry& Biology, vol. 5, No. 7, 365-372 (1998).
Nicolaou, K. C., et al., "Synthesis of Epothilones A and B in Solid and Solution Phase" (Correction to Nature 387, 268-272 (1997)), Nature, 390, 100 (1997).
Nicolaou, K. C., et al., "Synthesis of Epothilones A and B in Solid and Solution Phase", Nature, vol. 387, 268-272 (1997).
Nicolaou, K. C., et al., "Total Syntheses of Epothilone A: The Macrolactonization Approach", Angew. Chem. Int. Engl., vol. 36, No. 5, 525-527 (1997).
Nicolaou, K., et al., "Chemistry, Biology and Medicine of Selected Tubulin Polymerizing Agents", Pure Appl. Chem., vol. 71, No. 6, 989-997 (1999).
Nicolaou, K., et al., "Total Synthesis of Epothilone E and Related Side-chain Modified Analogues Via a Stille Coupling Based Strategy", Bioorg. Med. Chem., vol. 7, No. 5, 665-697 (1999).
Nicolaou, K.C., "Chemical Synthesis and Biological Properties of Pyridine Epothilones", Chemistry & Biology, vol. 7, No. 8, (2000) pp. 593-599.
Nicolaou, K.C., et al., "The Olefin Metathesis Approach to Epothilone A and Its Analogues", J. Am. Chem. Soc., vol. 119, No. 34, 7960-7973 (1997).
Nicolaou, K.C., et al., "Total Synthesis of Epothilones A and B via a Macrolactonization-Based Strategy", J. Am. Chem. Soc., vol. 119, No. 34, 7974-7991 (1997).
Raucher, S., et al., "Total Synthesis of (+)-Dihydrocostunolide via Tandem Cope-Claisen Rearrangement", J. Org. Chem., vol. 51, No. 26, 5503-5505 (1986).
Sato, M, et al., "Reduction of Organic Compounds with Low-Valent Niobium (NbCl5/NaAlH4)", Chem. Letters., 157-160 (1982).
Schinzer, D., et al., "Studies Towards the Total Synthesis of Epothilones: Asymmetric Synthesis of the Key Fragments", Chem. Eur. J., vol. 2, No. 22, 1477-1482 (1996).
Schinzer, D., et al., "Syntheses of (13)-Epothilone A", Chem. Eur. J., vol. 5, No. 9, 2483-2491 (1999).
Schinzer, D., et al., "Synthesis of (-)-Epothilone B", Chem. Eur. J., vol. 5, No. 9, 2492-2500 (1999).
Schinzer, D., et al., "Total Synthesis of (-)-Epothilone A", Angew. Chem. Int. Ed. Engl., vol. 36, No. 5, 523-524 (1997).
Schobert, R., et al., "Reduction and Isomerization of Oxiranes and alpha-Diazoketones by Various Early Transition Metallocenes", Synlett, vol. 8, 465-466 (1990).
Schobert, R., et al., "Reduction and Isomerization of Oxiranes and α-Diazoketones by Various Early Transition Metallocenes", Synlett, vol. 8, 465-466 (1990).
Sharpless, K.B., et al., "Lower Valent Tungsten Halides. A New Class of Reagents for Deoxygenation of Organic Molecules", J. Amer. Chem. Soc., vol. 94, No. 18, 6538-6540 (1972).
Su, D.-S., et al., "Structure-Activity Relationships of the Epothilones and the First In Vivo Comparison with Paclitaxel", Angew. Chem. Int. Ed. Eng.l, vol. 36, No. 19, 2093-2096 (1997).
Su, D.-S., et al., "Total Synthesis of (-)-Epothilone B: An Extension of the Suzuki Coupling Method and Insights into Structure-Activity Relationships of the Epothilones", Angew. Chem. Int. Ed. Engl., vol. 36, No. 7, 757-759 (1997).
Taylor, R. and Haley, J., "Towards the Synthesis of Epothilone A: Enantioselective Preparation of the Thiazole Sidechain and Macrocyclic Ring Closure", Tetrahedron Lett., vol. 38, No. 12, 2061-2064 (1997).
U.S. Appl. No. 08/856,533 filed May 14, 1997, K. Nicolaou et al.
U.S. Appl. No. 08/923,869 filed Sep. 4, 1997, K. Nicolaou et al.
U.S. Appl. No. 60/032,864 filed Dec. 13, 1996, K. Nicolaou et al.
U.S. Appl. No. 60/032,864, Nicolaou et al., filed Dec. 13, 1996.
Victory, S. F., et al., "Relative Stereochemistry and Solution Conformation of the Novel Paclitaxel-Like Antimitotic Agent Epothilone A", Bioorg. Med. Chem. Letts., vol. 6, No. 7, 893-898 (1996).
Winkler, J. D., et al., "A Model For The Taxol (Paclitaxel) Epothilone Pharmacophore", Bioorg. Med. Chem. Letters., vol. 6, No. 24, 2963-2966 (1966).
Yang, Z., et al., "Total Synthesis of Epothilone A: The Olefin Metathesis Approach", Angew. Chem. Int. Ed. Engl., vol. 36, No. 1/2, 166-168 (1997).

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2021142202A1 (en) 2020-01-10 2021-07-15 R-Pharm Us Operating Llc Compositions of ixabepilone

Also Published As

Publication number Publication date
BG108075A (en) 2005-04-30
RU2003128311A (en) 2005-03-10
US20020165258A1 (en) 2002-11-07
EP1385529A1 (en) 2004-02-04
CA2438610A1 (en) 2002-08-29
NO20033684D0 (en) 2003-08-19
WO2002066038A1 (en) 2002-08-29
US6686380B2 (en) 2004-02-03
HUP0303175A2 (en) 2003-12-29
BR0207487A (en) 2004-08-10
PL363362A1 (en) 2004-11-15
NO20033684L (en) 2003-10-13
KR20040025895A (en) 2004-03-26
MXPA03007394A (en) 2003-12-04
EE200300396A (en) 2003-12-15
IL156988A0 (en) 2004-02-08
EP1385529A4 (en) 2007-05-09
JP2004522774A (en) 2004-07-29
ZA200306173B (en) 2004-11-23
CN1774253A (en) 2006-05-17
IS6918A (en) 2003-08-18

Similar Documents

Publication Publication Date Title
USRE41393E1 (en) Treatment of refractory tumors using epothilone derivatives
US6727276B2 (en) Epothilone derivatives for the treatment of refractory tumors
AU2002248542B2 (en) Combination of epothilone analogs and chemotherapeutic agents for the treatment of proliferative diseases
CA2681962C (en) Methods of administering epothilone analogs for the treatment of cancer
AU2002248542A1 (en) Combination of epothilone analogs and chemotherapeutic agents for the treatment of proliferative diseases
EP1353668A1 (en) Methods of administering epothilone analogs for the treatment of cancer
AU2002245296B2 (en) Methods of administering epothilone analogs for the treatment of cancer
AU2002245296A1 (en) Methods of administering epothilone analogs for the treatment of cancer
AU2002247123A1 (en) Treatment of refractory tumors using epothilone derivatives
AU2002255539A1 (en) Epothilone derivatives for the treatment of refractory tumors
AU2002243548A1 (en) Parenteral formulation containing epothilone analogs

Legal Events

Date Code Title Description
FPAY Fee payment

Year of fee payment: 8

FPAY Fee payment

Year of fee payment: 12

AS Assignment

Owner name: R-PHARM US OPERATING LLC, NEW JERSEY

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:BRISTOL-MYERS SQUIBB COMPANY;REEL/FRAME:036585/0956

Effective date: 20150701