WO2007008496A1 - Formulations chimiotherapeutiques de trihydrochlorure de zosuquidar et cyclodextrines modifies - Google Patents

Formulations chimiotherapeutiques de trihydrochlorure de zosuquidar et cyclodextrines modifies Download PDF

Info

Publication number
WO2007008496A1
WO2007008496A1 PCT/US2006/026031 US2006026031W WO2007008496A1 WO 2007008496 A1 WO2007008496 A1 WO 2007008496A1 US 2006026031 W US2006026031 W US 2006026031W WO 2007008496 A1 WO2007008496 A1 WO 2007008496A1
Authority
WO
WIPO (PCT)
Prior art keywords
zosuquidar
sulfobutylcyclodextrin
cyclodextrin
stable
composition
Prior art date
Application number
PCT/US2006/026031
Other languages
English (en)
Inventor
Jeff Schwegman
Branimir Sikic
Daniel Hoth
David Socks
Scott Glenn
John Marcelletti
Michael J. Walsh
Pratik S. Multani
Original Assignee
Kanisa Pharmaceuticals, Inc.
Edgar, Mark
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from US11/416,829 external-priority patent/US20070010478A1/en
Priority claimed from US11/417,958 external-priority patent/US20070010485A1/en
Priority claimed from US11/416,992 external-priority patent/US20070009532A1/en
Priority claimed from US11/418,324 external-priority patent/US20070010486A1/en
Priority claimed from US11/418,400 external-priority patent/US20070010487A1/en
Application filed by Kanisa Pharmaceuticals, Inc., Edgar, Mark filed Critical Kanisa Pharmaceuticals, Inc.
Priority to EP06774486A priority Critical patent/EP1898957A1/fr
Priority to AU2006269498A priority patent/AU2006269498A1/en
Priority to CA002630087A priority patent/CA2630087A1/fr
Publication of WO2007008496A1 publication Critical patent/WO2007008496A1/fr

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • 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
    • A61K31/54Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one sulfur as the ring hetero atoms, e.g. sulthiame
    • A61K31/5415Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one sulfur as the ring hetero atoms, e.g. sulthiame ortho- or peri-condensed with carbocyclic ring systems, e.g. phenothiazine, chlorpromazine, piroxicam
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/08Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing oxygen, e.g. ethers, acetals, ketones, quinones, aldehydes, peroxides
    • A61K47/10Alcohols; Phenols; Salts thereof, e.g. glycerol; Polyethylene glycols [PEG]; Poloxamers; PEG/POE alkyl ethers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/16Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing nitrogen, e.g. nitro-, nitroso-, azo-compounds, nitriles, cyanates
    • A61K47/18Amines; Amides; Ureas; Quaternary ammonium compounds; Amino acids; Oligopeptides having up to five amino acids
    • A61K47/183Amino acids, e.g. glycine, EDTA or aspartame
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/26Carbohydrates, e.g. sugar alcohols, amino sugars, nucleic acids, mono-, di- or oligo-saccharides; Derivatives thereof, e.g. polysorbates, sorbitan fatty acid esters or glycyrrhizin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/30Macromolecular organic or inorganic compounds, e.g. inorganic polyphosphates
    • A61K47/36Polysaccharides; Derivatives thereof, e.g. gums, starch, alginate, dextrin, hyaluronic acid, chitosan, inulin, agar or pectin
    • A61K47/40Cyclodextrins; Derivatives thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/14Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles
    • A61K9/19Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles lyophilised, i.e. freeze-dried, solutions or dispersions

Definitions

  • the present invention relates to a method of treating patients with leukemias, solid tumors, and other malignancies using chemotherapeutic agents in combination with zosuquidar that has been solubilized by a modified cyclodextrin, such as sulfobutylcyclodextrin or hydroxypropyl cyclodextrin.
  • the invention is also directed to pharmaceutical formulations comprising zosuquidar in combination with a modified cyclodextrin.
  • Drug resistance is one of the most difficult problems that must be overcome in order to achieve successful treatment of human tumors with chemotherapy.
  • drug resistance a characteristic of intrinsically resistant tumors (for example, colon, renal, and pancreas) or other malignancies, may be evident at the onset of therapy.
  • acquired drug resistance results when tumors or malignancies initially respond to therapy but become refractory to subsequent treatments. Once a tumor or malignancy has acquired resistance to a specific chemotherapeutic agent, it is common to observe collateral resistance to other structurally similar agents.
  • Multidrug resistance the ability of cancer cells to become resistant to the agent(s) actively used for therapy, as well as other drugs that are structurally and functionally unrelated, is a particularly insidious form of drug resistance.
  • Zosuquidar a 10,11-methanobenzosuberane derivative, is useful in enhancing the efficacy of existing cancer chemotherapeutics and for treating multidrug resistance.
  • zosuquidar has limited solubility in aqueous solution, such that the formulation concentration is limited, resulting in a large number of vials to contain doses in the potentially efficacious range (e.g., a clinical formulation of zosuquidar without solubility enhancers of 50 mg per 30 mL vial that requires 11 units to provide 550 mg of zosuquidar).
  • Dosage forms and treatment regimens for treating solid tumors leukemias such as acute myelogenous leukemia (AML) and other malignancies that result in increased rates of complete remission and increased cancer-free survival rates are desirable.
  • intravenous zosuquidar formulations having a greater zosuquidar concentration and increased content per dosage unit.
  • Zosuquidar formulated with a modified cyclodextrin to enhance its solubility provides an improved formulation that can offer such advantages. Hydroxypropylcyclodextrins and sulfobutylcyclodextrins are particularly preferred modified cyclodextrins for use in zosuquidar formulations.
  • a stable chemotherapeutic composition comprising zosuquidar in combination with a modified cyclodextrin.
  • the modified cyclodextrin is a hydroxypropyl- ⁇ -cyclodextrin.
  • the modified cyclodextrin is a sulfobutylcyclodextrin, e.g., a polyanionic ⁇ -cyclodextrin derivative with a sodium sulfonate salt separated from a lipophilic cavity by a butyl ether spacer group.
  • the composition is in lyophilized form.
  • the composition is in solution form.
  • the composition is in liquid unit dosage form, comprising from about 10 mg/mL to about 30 mg/mL zosuquidar and from about 100 mg/mL to about 200 mg/mL sulfobutylcyclodextrin.
  • the composition is in liquid unit dosage form, comprising from about 20 mg/mL to about 25 mg/mL zosuquidar and from about 125 mg/mL to about 175 mg/mL sulfobutylcyclodextrin.
  • the composition is in liquid unit dosage form, comprising about 22.5 mg/mL zosuquidar and about 150 mg/mL sulfobutylcyclodextrin.
  • the composition is in lyophilized form, comprising zosuquidar and sulfobutylcyclodextrin in a weight ratio of zosuquidar to sulfobutylcyclodextrin of from about 1:5.7 to about 1:7.4.
  • the composition is in lyophilized form, comprising zosuquidar and sulfobutylcyclodextrin in a weight ratio of zosuquidar to sulfobutylcyclodextrin of from about 1:6 to about 1:7.
  • the composition is in lyophilized form, comprising zosuquidar and sulfobutylcyclodextrin in a weight ratio of zosuquidar to sulfobutylcyclodextrin of about 1 :6.73.
  • the solution is a dextrose solution.
  • a pharmaceutical kit comprising at least one container containing a stable chemotherapeutic composition comprising zosuquidar in combination with a modified cyclodextrin; and directions for administering the chemotherapeutic composition to treat a malignancy that expresses P-glycoprotein.
  • the modified cyclodextrin is hydroxypropyl- ⁇ -cyclodextrin.
  • the modified cyclodextrin is sulfobutylcyclodextrin.
  • the malignancy is acute myelogenous leukemia.
  • the kit further comprises at least one container containing daunorubicin and at least one container containing cytarabine, and directions for administering the daunorubicin and cytarabine to treat newly diagnosed acute myelogenous leukemia.
  • the kit further comprises at least one container containing Mylotarg, and directions for administering the Mylotarg to treat relapsed acute myelogenous leukemia.
  • a pharmaceutical kit comprising at least one vial containing a stable chemotherapeutic lyophilized composition, comprising about 275 mg/vial zosuquidar and about 1850 mg/vial sulfobutylcyclodextrin; and directions for reconstituting the lyophilized composition with a 15 mL of a 5% dextrose solution and administering the reconstituted solution to a patient to treat acute myelogenous leukemia.
  • a method of treating cancer in a patient exhibiting positive P- glycoprotein expression or positive P-glycoprotein function comprising administering to the patient a chemotherapeutic agent that is a substrate for P- glycoprotein efflux and a stable chemotherapeutic composition comprising zosuquidar in combination with a modified cyclodextrin, whereby the cancer is treated.
  • the modified cyclodextrin is a hy droxypropyl- ⁇ -cyclodextrin.
  • the modified cyclodextrin is a sulfobutylcyclodextrin, e.g., a polyanionic ⁇ -cyclodextrin derivative with a sodium sulfonate salt separated from a lipophilic cavity by a butyl ether spacer group.
  • the stable chemotherapeutic composition is in lyophilized form.
  • the stable chemotherapeutic composition is in solution form.
  • the stable chemotherapeutic composition is in liquid unit dosage form, comprising from about 10 mg/mL to about 30 mg/mL zosuquidar and from about 100 mg/mL to about 200 mg/mL sulfobutylcyclodextrin. In an embodiment of the fourth aspect, the stable chemotherapeutic composition is in liquid unit dosage form, comprising from about 20 mg/mL to about 25 mg/mL zosuquidar and from about 125 mg/mL to about 175 mg/mL sulfobutylcyclodextrin.
  • the stable chemotherapeutic composition is in liquid unit dosage form, comprising about 22.5 mg/mL zosuquidar and about 150 mg/mL sulfobutylcyclodextrin.
  • the stable chemotherapeutic composition is in lyophilized form, comprising zosuquidar and sulfobutylcyclodextrin in a weight ratio of zosuquidar to sulfobutylcyclodextrin of from about 1 :5.7 to about 1 :7.4.
  • the stable chemotherapeutic composition is in lyophilized form, comprising zosuquidar and sulfobutylcyclodextrin in a weight ratio of zosuquidar to sulfobutylcyclodextrin of from about 1 :6 to about 1 :7.
  • the stable chemotherapeutic composition is in lyophilized form, comprising zosuquidar and sulfobutylcyclodextrin in a weight ratio of zosuquidar to sulfobutylcyclodextrin of about 1 :6.73.
  • the stable chemotherapeutic composition is a dextrose solution.
  • the cancer is acute myelogenous leukemia.
  • the cancer is a carcinoma, e.g., breast cancer or ovarian cancer.
  • the cancer is a sarcoma.
  • the cancer is a hematologic malignancy, e.g., acute lymphoblastic leukemia, chronic myeloid leukemia, plasma cell dyscrasias, lymphoma, and myelodysplasia.
  • the chemotherapeutic agent is an anthracycline, e.g., doxorubicin, daunorubicin, epirubicin, idarubicin, or mitoxantrone.
  • the chemotherapeutic agent is a Topoisomerase-II inhibitor, e.g., etoposide or teniposide.
  • the chemotherapeutic agent is a vinca, e.g., vincristine, vinblastine, vinorelbine, or vindesine.
  • the chemotherapeutic agent is a taxane, e.g., paclitaxel or docetaxel.
  • the chemotherapeutic agent is selected from the group consisting of gleevec, dactinomycin, bisantrene, mitoxantrone, actinomyocin D, mithomycin C, mitramycin, methotrexate, adriamycin, mitomycin, mithramycin, anthracene, and epipodophyllo-toxin.
  • the chemotherapeutic agent comprises daunorubicin and cytarabine, and the cancer is newly diagnosed acute myelogenous leukemia.
  • the chemotherapeutic agent comprises Mylotarg
  • the cancer is relapsed acute myelogenous leukemia.
  • a method of administering a therapeutic agent that is a substrate for P-glycoprotein efflux to a patient in need thereof is provided, wherein the patient exhibits positive P-glycoprotein expression or P-glycoprotein function, the method comprising administering the therapeutic agent to the patient; and administering a stable P-glycoprotein efflux pump inhibiting composition comprising zosuquidar in combination with a modified cyclodextrin to the patient.
  • the modified cyclodextrin is a hydroxypropyl- ⁇ -cyclodextrin.
  • the modified cyclodextrin is a sulfobutylcyclodextrin, e.g., a polyanionic ⁇ -cyclodextrin derivative with a sodium sulfonate salt separated from a lipophilic cavity by a butyl ether spacer group.
  • the stable chemotherapeutic composition is in lyophilized form.
  • the stable chemotherapeutic composition is in solution form. In an embodiment of the fifth aspect, the stable chemotherapeutic composition is in liquid unit dosage form, comprising from about 10 mg/mL to about 30 mg/mL zosuquidar and from about 100 mg/mL to about 200 mg/mL sulfobutylcyclodextrin.
  • the stable chemotherapeutic composition is in liquid unit dosage form, comprising from about 20 mg/mL to about 25 mg/mL zosuquidar and from about 125 mg/mL to about 175 mg/mL sulfobutylcyclodextrin.
  • the stable chemotherapeutic composition is in liquid unit dosage form, comprising about 22.5 mg/mL zosuquidar and about 150 mg/mL sulfobutylcyclodextrin.
  • the stable chemotherapeutic composition is in lyophilized form, comprising zosuquidar and sulfobutylcyclodextrin in a weight ratio of zosuquidar to sulfobutylcyclodextrin of from about 1 :5.7 to about 1 :7.4.
  • the stable chemotherapeutic composition is in lyophilized form, comprising zosuquidar and sulfobutylcyclodextrin in a weight ratio of zosuquidar to sulfobutylcyclodextrin of from about 1 :6 to about 1 :7.
  • the stable chemotherapeutic composition is in lyophilized form, comprising zosuquidar and sulfobutylcyclodextrin in a weight ratio of zosuquidar to sulfobutylcyclodextrin of about 1 :6.73.
  • the stable chemotherapeutic composition is a dextrose solution.
  • the therapeutic agent comprises an immunosuppressant, e.g., cyclosporine, cyclosporine A, and tacrolimus.
  • an immunosuppressant e.g., cyclosporine, cyclosporine A, and tacrolimus.
  • the therapeutic agent comprises a steroid, e.g., dexamethasone, hydrocortisone, corticosterone, triamcinolone, aldosterone, and methy lprednisolone .
  • a steroid e.g., dexamethasone, hydrocortisone, corticosterone, triamcinolone, aldosterone, and methy lprednisolone .
  • the therapeutic agent comprises an antiepileptic, e.g., phenytoin.
  • the therapeutic agent comprises an antidepressant, e.g., citalopram, thioperidone, trazodone, trimipramine, amitriptyline, and phenothiazines.
  • an antidepressant e.g., citalopram, thioperidone, trazodone, trimipramine, amitriptyline, and phenothiazines.
  • the therapeutic agent comprises an antipsychotic, e.g., fluphenazine, haloperidol, thioridazine, and trimipramine.
  • an antipsychotic e.g., fluphenazine, haloperidol, thioridazine, and trimipramine.
  • the therapeutic agent comprises a protease inhibitor, e.g., amprenavir, indinavir, lopinavir, nelfmavir, ritonavir, and saquinavir.
  • a protease inhibitor e.g., amprenavir, indinavir, lopinavir, nelfmavir, ritonavir, and saquinavir.
  • the therapeutic agent comprises a calcium blocker, e.g., bepridil, diltiazem, flunarizine, lomerizine, secoverine, tamolarizine, verapamil, nicardipine, prenylamine, and fendiline.
  • a calcium blocker e.g., bepridil, diltiazem, flunarizine, lomerizine, secoverine, tamolarizine, verapamil, nicardipine, prenylamine, and fendiline.
  • the therapeutic agent comprises a cardiac drug, e.g., digoxin, diltiazem, verapamil, and talinolol.
  • a cardiac drug e.g., digoxin, diltiazem, verapamil, and talinolol.
  • the therapeutic agent comprises daunorubicin and cytarabine, and the patient is newly diagnosed with acute myelogenous leukemia.
  • the therapeutic agent comprises Mylotarg, and the patient is diagnosed with relapsed acute myelogenous leukemia.
  • Figure 1 illustrates the increase of zosuquidar concentration in solution as a function of sulfobutylcyclodextrin concentration.
  • U.S. Pat. Nos. 5,643,909 and 5,654,304 disclose a series of 10,11- methanobenzosuberane derivatives useful in enhancing the efficacy of existing cancer chemotherapeutics and for treating multidrug resistance.
  • One such derivative having good activity, oral bioavailability, and stability, is zosuquidar, a compound of formula (2R)-anti-5-
  • PK pharmacokinetic
  • the zosuquidar employed in formulations of preferred embodiments can be administered in the form of a pharmaceutically acceptable salt, e.g., the trihydrochloride salt.
  • a pharmaceutically acceptable salt e.g., the trihydrochloride salt.
  • pharmaceutically acceptable salts and “a pharmaceutically acceptable salt thereof as used herein in regard to therapeutic agents are broad terms and are used in their ordinary sense, including, without limitation, to refer to salts prepared from pharmaceutically acceptable, non-toxic acids ⁇ e.g., as for zosuquidar) or bases (for other therapeutic agents capable of forming a salt with a base).
  • Suitable pharmaceutically acceptable salts include metallic salts, e.g., salts of aluminum, zinc, alkali metal salts such as lithium, sodium, and potassium salts, alkaline earth metal salts such as calcium and magnesium salts; organic salts, e.g., salts of organic acids ⁇ e.g., benzenesulfonate, mesylate, fumarate, citrate), lysine, N,N'-dibenzylethylenediamine, chloroprocaine, choline, diethanolamine, ethylenediamine, meglumine (N-methylglucamine), procaine, and tris; salts of free acids and bases; inorganic salts, e.g., sulfate, hydrochloride, and hydrobromide; and other salts which are currently in widespread pharmaceutical use and are listed in sources well known to those of skill in the art, such as, for example, The Merck Index.
  • metallic salts e.g., salts of aluminum, zinc, alkal
  • Any suitable constituent can be selected to make a salt of zosuquidar or other therapeutic agents discussed herein, provided that it is non-toxic and does not substantially interfere with the desired activity.
  • pharmaceutically acceptable precursors and derivatives of the compounds can be employed.
  • Pharmaceutically acceptable amides, lower alkyl esters, protected derivatives, and chelates can also be suitable for use in compositions and methods of preferred embodiments.
  • Also suitable for administration are selected therapeutic agents in hydrated form, selected enantiomeric forms of certain therapeutic agents, racemic mixtures of certain therapeutic agents, and the like.
  • Zosuquidar is generally administered in the form of the trihydrochloride salt.
  • Conventional zosuquidar trihydrochloride formulations include those containing zosuquidar (50 mg as free base), glycine (15 mg), and mannitol (200 mg) dissolved in enough water for injection, to yield a free base concentration of 5 mg/mL.
  • the formulation is filled into vials and lyophilized to give a vial containing 50 mg of free base.
  • a 30 mL vial size is necessary to contain 50 mg of the zosuquidar formulation.
  • For a typical >200 mg dose of zosuquidar multiple 50 mg vials are needed to contain the formulation, greatly increasing manufacturing costs and reducing convenience for the end user ⁇ e.g., a pharmacist).
  • Modified Cyclodextrins Modified Cyclodextrins
  • Cyclodextrins are cyclic oligomers of glucose; these compounds form inclusion complexes with any drug whose molecule can fit into the lipophile-seeking cavities of the cyclodextrin molecule. See U.S. Pat. No. 4,727,064 for a description of various cyclodextrin derivatives. Cyclodextrins of preferred embodiments can include ⁇ -, ⁇ -, and ⁇ -cyclodextrins. The ⁇ -cyclodextrins include six glucopyranose units, the ⁇ - cyclodextrins include seven glucopyranose units, and the ⁇ -cyclodextrins include eight glucopyranose units.
  • the ⁇ -cyclodextrins are generally preferred as having a suitable cavity size for zosuquidar.
  • Cyclodextrin can be in any suitable form, including amorphous and crystalline forms, with the amorphous form generally preferred.
  • Cyclodextrins suitable for use in the formulations of preferred embodiments include the hydroxypropyl, hydroxyethyl, glucosyl, maltosyl, and maltotrosyl derivatives of ⁇ - cyclodextrin, carboxyamidomethyl- ⁇ -cyclodextrin, carboxymethyl- ⁇ -cyclodextrin, and diethylamino- ⁇ -cyclodextrin.
  • Chemically modified and substituted ⁇ -, ⁇ -, and ⁇ -cyclodextrins are generally preferred over unmodified ⁇ -, ⁇ -, and ⁇ -cyclodextrins due to improved toxicity and solubility properties.
  • the degree of substitution of the hydroxy 1 groups of the glucopyranose units of the cyclodextrin ring can affect solubility. In general, a higher average degree of substitution of substituent groups in the cyclodextrin molecule yields a cyclodextrin of higher solubility.
  • a stable association is necessary.
  • non-covalent forces such as van der Waal forces, hydrophobic interaction, dipole moment and other forces are responsible for formation of a stable complex.
  • more than one guest molecule may fit into the cavity.
  • more than one molecule of cyclodextrin might bind to the guest molecule. Only a portion of the molecule must fit into the cavity to form a complex.
  • a one-to-one molar ratio is not always achieved, especially with high or low molecular weight guest molecules.
  • the guest molecule associates with the cyclodextrin so that the hydrophobic portion of the guest interacts with the hydrophobic cavity of the cyclodextrin. This interaction is an equilibrium reaction, with the direction of the equilibrium dependent upon the guest molecule. For some guest molecules, the complex is predominant while for other guest molecules, the free state might be preferred. In order to reduce the probability of free guest molecules self-associating to form an insoluble precipitate, excess cyclodextrin is frequently used to increase the probability of the guest molecule associating with the cavity of the cyclodextrin rather than associating with other guest molecules.
  • a moderate excess of the cyclodextrin is generally desirable.
  • a molar ratio of zosuquidar to the cyclodextrin approaching one-to-one may be preferred.
  • Sulfobutyl- ⁇ -cyclodextrin is a particularly preferred modified cyclodextrin for solubilizing zosuquidar.
  • This cyclodextrin is marketed by CyDex, Inc., (Lenexa, KS) under the trade name CAPTISOL®.
  • CAPTISOL® cyclodextrins are polyanionic ⁇ - cyclodextrin derivatives with a sodium sulfonate salt separated from the lipophilic cavity by a butyl ether spacer group, or sulfobutylether (SBE).
  • Sulfobutylcyclodextrin may provide a beneficial and protected environment for zosuquidar in its lipophilic cavity while its hydrophilic surface contributes good water solubility, improving both solubility and stability. Interaction of the zosuquidar with sulfobutylcyclodextrin may reduce decomposition by protecting the labile region from potential reactants in the aqueous environment. The inherent pharmacokinetics and pharmacodynamics of zosuquidar are unaffected by sulfobutylcyclodextrin.
  • Hydroxypropylcyclodextrin Hydroxypropyl- ⁇ -cyclodextrin is also a preferred modified cyclodextrin for solubilizing zosuquidar. This cyclodextrin is marketed by RDI Division of Fitzgerald Industries Intl., (Concord, MA). Hydroxypropyl- ⁇ -cyclodextrin is produced from ⁇ - cyclodextrin by hydroxpropylation of the hydroxyl groups of the cyclodextrin.
  • the basic closed circular structure of ⁇ -cyclodextrin is maintained in hydroxypropyl- ⁇ -cyclodextrin.
  • the glycosidic oxygen forming the bond between the adjacent glucose monomers and the hydrogen atoms lining the cavity of the cyclodextrin impart an electron density and hydrophobic character to the cavity.
  • Organic compounds, such as zosuquidar, interact with the walls of the cavity to form inclusion complexes.
  • the hydroxyl groups and the hydroxypropyl groups are on the exterior of the molecule and interact with water to provide the increased aqueous solubility of the hydroxypropyl- ⁇ - cyclodextrin and the complexes made with the hydroxypropyl- ⁇ -cyclodextrin.
  • the hydroxypropyl groups are randomly substituted onto the hydroxyl groups of the ⁇ -cyclodextrin and the amount of substitution is reported as average degree of substitution or number of hydroxypropyl groups per ⁇ -cyclodextrin.
  • some molecules will have more substituents than the average number of substituents and some less.
  • the result is a mixture of many molecules varying with respect to the number and location of substitutions around the ring of the ⁇ - cyclodextrin.
  • Substitution can have an effect on the binding of guest molecules to the hydroxypropyl- ⁇ -cyclodextrin. At low degrees of substitution, binding is very similar to that of the unmodified B-cyclodextrin.
  • a preferred average degree of substitution of hydroxypropyl- ⁇ -cyclodextrin when employed in combination with zosuquidar is from about 4 or 5 to about 6, 7, or 8.
  • Hydroxypropyl- ⁇ -cyclodextrin is very soluble in water, with substitution of the hydroxy 1 groups of the ⁇ -cyclodextrin disrupting the network of hydrogen bonding around the rim of the ⁇ -cyclodextrin. As a result of disruption of the hydrogen-bonding network, the hydroxyl groups interact much more strongly with water, resulting in increased solubility compared to ⁇ -cyclodextrin. Hydroxypropyl- ⁇ -cyclodextrin is generally more soluble than unmodified ⁇ -cyclodextrin. For hydroxypropyl- ⁇ -cyclodextrin having a degree of substitution of 7.6, the solubility in aqueous solution is 360g/100ml.
  • Hydroxypropyl- ⁇ -cyclodextrin is also soluble in aqueous ethanol (225g/100ml for a 95% ethanol solution). In preferred formulations, the solubility of the complex with zosuquidar is not generally exceeded. Complexes of zosuquidar and hydroxypropyl- ⁇ - cyclodextrins exhibit increased solubility and stability when compared to corresponding complexes of zosuquidar and unmodified ⁇ -cyclodextrins.
  • Strong acids such as hydrochloric acids, can hydrolyze hydroxypropyl- ⁇ - cyclodextrin.
  • the rate of hydrolysis is dependent upon the temperature and concentration of the acid. The higher the temperature or concentration of the acid, the more rapid is the rate of hydrolysis.
  • Weak acids such as organic acids, do not hydrolyze hydroxypropyl- ⁇ - cyclodextrin, and hydroxypropyl- ⁇ -cyclodextrin is stable in bases.
  • Hydroxypropyl- ⁇ - cyclodextrin is not hydrolyzed by ⁇ -amylase or glucoamylase, but ⁇ -cyclodextrin can be hydrolyzed by some ⁇ -amylases. Hydroxypropyl- ⁇ -cyclodextrin generally exhibits good stability under physiological conditions when employed in formulations for intravenous use.
  • zosuquidar sulfobutylcyclodextrin formulations
  • suitable cyclodextrins such as hydroxypropyl- ⁇ -cyclodextrins
  • sulfobutylcyclodextrin can be used instead of sulfobutylcyclodextrin to solubilize zosuquidar.
  • a mixture of two or more different cyclodextrins can be used.
  • sulfobutylcyclodextrin formulation (lyophilized) allows an 800 mg dose of zosuquidar to be contained in one (50 niL vial) or two vials (20 or 30 mL vial) versus three 100 mL vials for a zosuquidar formulation without cyclodextrin, resulting in greater manufacturing efficiency.
  • the relative amounts of zosuquidar and the cyclodextrin, e.g., sulfobutylcyclodextrin can be adjusted, depending upon the particular formulation and the specific cyclodextrin employed.
  • a molar ratio of zosuquidar to modified cyclodextrin of from about 1:1 or less to about 1 :10 or more is generally preferred, preferably from about 1 :5.0 or 1 :5.5 to about 1 :8.0, 1:8.5, 1 :9.0, or 1 :9.5, and more preferably from about 1:5.7, 1:5.8, 1:5.9, 1:6.0, 1:6.1, 1:6.2, 1:6.3, 1:6.4, 1:6.5, 1:6.6, 1:6.7 to about 1 :6.8, 1 :6.9, 1:7.0, 1 :7.1, 1 :7.2, 1:7.3, or 1.7:4.
  • the zosuquidar - modified cyclodextrin formulation can by supplied as a powder and reconstituted. Alternatively, it can be provided in the form of an aqueous liquid, which can optionally be freeze dried or lyophilized.
  • the zosuquidar - modified cyclodextrin formulations are prepared by dissolving the cyclodextrin in water and adding the zosuquidar to the aqueous modified cyclodextrin solution. Excipients, if any are desired may be added with or subsequent to adding the active compound. The resulting solution can be sterilized using any of the known methods appropriate to preserving the active compound.
  • the components can be sterilized by any of the known methods appropriate to preserving zosuquidar prior to mixing in water and can be mixed using sterile equipment and techniques.
  • the solution can be lyophilized in sterile containers and capped.
  • the lyophilized composition of matter Prior to use, can be reconstituted using sterile water for injection, deionized sterilized water, 5% dextrose solution, or other appropriate diluent.
  • Contemplated routes of administration include topical, oral, subcutaneous, parenteral, intradermal, intramuscular, intraperitoneal, and intravenous. However, it is particularly preferred to administer the zosuquidar - modified cyclodextrin in intravenous form.
  • the intravenous forms containing zosuquidar - modified cyclodextrin are preferably isotonic with the blood or other body fluid of the patient.
  • the isotonicity of the compositions can be attained using sodium tartrate, propylene glycol, sodium chloride, or other inorganic or organic solutes.
  • Buffering agents can be employed, such as acetic acid and salts, citric acid and salts, boric acid and salts, and phosphoric acid and salts.
  • Parenteral vehicles include, Ringer's dextrose, lactated Ringer's, or fixed oils.
  • Intravenous vehicles can include fluid and nutrient replenishers, electrolyte replenishers (such as those based on Ringer's dextrose), and the like.
  • a particularly preferred vehicle is dextrose solution, e.g., 5% dextrose.
  • Various excipients can be employed, depending upon the route of administration and the preparation desired. Standard texts, such as “Remington: The Science and Practice of Pharmacy”, Lippincott Williams & Wilkins; 20th edition (June 1, 2003) and “Remington's Pharmaceutical Sciences,” Mack Pub.
  • excipients which can include additional complexing agents, metal ions, polymeric compounds such as polyacetic acid, polyglycolic acid, hydrogels, dextran, and the like, liposomes, microemulsions, micelles, unilamellar or multilamellar vesicles, erythrocyte ghosts or spheroblasts.
  • Suitable lipids for liposomal formulation include, without limitation, monoglycerides, diglycerides, sulfatides, lysolecithin, phospholipids, saponin, bile acids, and the like.
  • the presence of such additional components can influence the physical state, solubility, stability, rate of in vivo release, and rate of in vivo clearance, and are thus chosen according to the intended application, such that the characteristics of the carrier are tailored to the selected route of administration.
  • a pharmaceutically acceptable preservative can be employed to increase the shelf life of the pharmaceutical compositions.
  • Benzyl alcohol can be suitable, although a variety of preservatives including, for example, parabens, thimerosal, chlorobutanol, or benzalkonium chloride can also be employed.
  • a suitable concentration of the preservative is typically from about 0.02% to about 2% based on the total weight of the composition, although larger or smaller amounts can be desirable depending upon the agent selected.
  • the zosuquidar — modified cyclodextrin complex can be provided to an administering physician or other health care professional in the form of a kit.
  • the kit is a package which houses one or more containers which contain zosuquidar complexed with a modified cyclodextrin, such as sulfobutylcyclodextrin or hydroxypropyl- ⁇ -cyclodextrin, in a suitable form and instructions for reconstituting and/or administering the pharmaceutical composition to a subject.
  • the kit can optionally also contain one or more additional therapeutic agents, e.g., mylotarg, daunorubicin, cytarabine, and/or other chemotherapeutic agents.
  • the kit can optionally contain one or more diagnostic tools and instructions for use.
  • kits containing a single composition comprising a complex of zosuquidar and sulfobutylcyclodextrin or hydroxypropyl- ⁇ -cyclodextrin in combination with one or more additional therapeutic agents can be provided, or separate pharmaceutical compositions containing a complex of zosuquidar - sulfobutylcyclodextrin and additional therapeutic agents can be provided.
  • the kit can also contain separate doses of zosuquidar - sulfobutylcyclodextrin complex for serial or sequential administration.
  • the kit can contain suitable delivery devices, e.g., syringes and the like, along with instructions for administrating the complex and any other therapeutic agent.
  • kits can optionally contain instructions for storage, reconstitution (if applicable), and administration of any or all therapeutic agents included.
  • the kits can include a plurality of containers reflecting the number of administrations to be given to a subject.
  • a kit for the treatment of a leukemia or solid tumor is provided.
  • a kit for the treatment of acute myelogenous leukemia is provided that includes a zosuquidar - sulfobutylcyclodextrin complex and mylotarg (for relapsed patients) or daunorubicin and cytarabine (for newly-diagnosed patients) and instructions for administering each.
  • a kit for the treatment of acute myelogenous leukemia includes a zosuquidar - sulfobutylcyclodextrin complex and one or more diagnostics or instructions for conducting one or more diagnostics for determining P-gp expression and/or efflux pump activity.
  • the kit can also include instructions, an assay, or a diagnostic for determining if a patient has acute myelogenous leukemia.
  • the kit can contain suitable delivery devices, e.g., syringes, inhalation devices, and the like, along with instructions for administrating zosuquidar and/or other therapeutic agent.
  • the kit can optionally contain instructions for storage, reconstitution (if applicable, e.g., for a lyophilized form reconstituted for intravenous administration), and administration of any or all therapeutic agents included.
  • the kits can include a plurality of containers reflecting the number of administrations to be given to a subject.
  • Contemplated amounts of solubilized zosuquidar for intravenous administration are from about 400 mg/day of zosuquidar or less to about 1,600 mg/day zosuquidar or more, preferably from about 500 or 600 mg/day to about 800, 900, 1000, 1 100, 1200, 1300, 1400, or 1500 mg/day, and most preferably 700 mg/day.
  • the duration of the injection of the zosuquidar - modified cyclodextrin complex can be adjusted depending upon various factors, and can comprise a single injection administered over the course of a few seconds or less to 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 26, 28, 30, 32, 34, 36, 40, 44, 48, 54, 60, 66, 72, 78, 84, 90, or 96 hours or more of continuous intravenous administration.
  • the zosuquidar - sulfobutylcyclodextrin complex formulations of preferred formulations are useful therapeutic agents for treating multidrug resistance in patients treated for malignancies, solid tumors, and leukemias.
  • the formulations are useful for treatment of cancers that express P-gp, e.g., many solid tumors, bladder cancer, pancreatic cancer, liver cancer, myeloma, carcinomas (e.g., breast cancer and ovarian cancer), sarcomas, and hematologic malignancies (e.g., acute myelogenous leukemia, acute lymphoblastic leukemia, chronic myeloid leukemia, plasma cell dyscrasias, lymphoma, myelodysplasia).
  • P-gp e.g., many solid tumors, bladder cancer, pancreatic cancer, liver cancer, myeloma, carcinomas (e.g., breast cancer and ovarian cancer), sarcomas, and he
  • the zosuquidar — sulfobutylcyclodextrin formulations are suitable for use in conjunction with suitable chemotherapeutic agents used to treat malignancies wherein multidrug resistance is of concern. However, the formulations are particularly suited for use in treating acute myelogenous leukemia.
  • relapsed patients are treated with mylotarg in combination with zosuquidar - sulfobutylcyclodextrin complex formulations.
  • Newly-diagnosed patients can be treated with daunorubicin and cytarabine in combination with zosuquidar - sulfobutylcyclodextrin complex formulations.
  • chemotherapeutic agents can also be used in combination with the zosuquidar - sulfobutylcyclodextrin complex formulations of preferred embodiments, e.g., anthracyclines (e.g., doxorubicin, daunorubicin, epirubicin, idarubicin, mitoxantrone), vincas (e.g., vincristine, vinblastine, vinorelbine, vindesine), Topoisomerase-II (e.g., etoposide, teniposide), taxanes (e.g., paclitaxel, docetaxel), and others (e.g., Gleevec, Mylotarg, dactinomycin, mithramycin).
  • anthracyclines e.g., doxorubicin, daunorubicin, epirubicin, idarubicin, mitoxantrone
  • vincas e.g
  • a P-gp expression or efflux pump activity diagnostic is conducted to provide information in treating AML patients or patients with metastatic breast cancer with a zosuquidar - cyclodextrin complex (e.g., zosuquidar - sulfobutylcyclodextrin or zosuquidar - hydroxypropyl cyclodextrin) in combination with Mylotarg.
  • a zosuquidar - cyclodextrin complex e.g., zosuquidar - sulfobutylcyclodextrin or zosuquidar - hydroxypropyl cyclodextrin
  • results of the P-gp expression or efflux pump activity diagnostic indicates positive P-gp expression or efflux pump activity, then treatment with a zosuquidar - cyclodextrin complex in combination with Mylotarg is initiated. If the results of the P-gp expression or efflux pump activity diagnostic indicate negative P-gp expression or efflux pump activity, then zosuquidar is expected not to yield an improvement in clinical outcome and another treatment option not involving administration of a P-gp efflux inhibitor is selected. In relapsed AML patients, it is generally considered acceptable clinical practice to wait for P-gp expression or efflux pump activity test results before initiating a treatment.
  • P-gp expression or efflux pump activity of a sample both in the presence and absence of the P-gp efflux inhibitor is compared, whereby the P-gp efflux that is inhabitable by the P- gp efflux inhibitor can be determined.
  • P-gp expression or function status correlates with expectation of clinical success, it can be useful to determine P-gp expression or efflux pump activity at any point in time.
  • Mylotarg was approved in May 2000 for relapsed CD33 -positive AML patients over the age of 60.
  • Mylotarg from Wyeth and Celltech is based on antibody-targeted chemotherapy.
  • Mylotarg' s highly specific antibody recognizes a cell-surface molecule, CD33, which is abundant on AML cells (>90%) but absent from normal blood stem cells, the seeds from which normal blood and immune cells originate.
  • the antibody is linked to calicheamicin, a potent chemotherapy agent. The antibody selectively targets leukemic blast cells and delivers calicheamicin to them.
  • the chemical structure of Mylotarg is provided below.
  • zosuquidar a highly specific and safe P-gp efflux inhibitor, complexed with cyclodextrin, in combination with Mylotarg or another calicheamicin- antibody conjugate is effective for treatment of relapsed AML.
  • the effective dose of the zosuquidar - cyclodextrin complex and the timing of administration of zosuquidar and Mylotarg are critical to achieving improved complete remission rates and enhanced leukemia free and overall survival rates in the relapsed AML patient population. While the methods and formulations of preferred embodiments are especially preferred for treatment of relapsed AML patients, the methods and formulations can be adapted to other drugs and indications.
  • P-gp efflux inhibitors other than Mylotarg can be administered according to the disclosed dosing regimens, or slightly modified dosing regimens.
  • the formulations and dosing regimens employing a zosuquidar - cyclodextrin complex and Mylotarg can be employed in treating AML patients other than relapsed AML patients, or for other types of leukemia or other cancers that express P-gp, e.g., many solid tumors, lymphomas, bladder cancer, pancreatic cancer, ovarian cancer, liver cancer, myeloma, lymphocytic leukemia, breast cancer, and sarcoma.
  • the duration of the injection of a zosuquidar- cyclodextrin complex and/or Mylotarg can be adjusted depending upon various factors, and can comprise a single injection administered over the course of a few seconds or less to 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 26, 28, 30, 32, 34, 36, 40, 44, 48, 54, 60, 66, 72, 78, 84, 90, or 96 hours or more of continuous intravenous administration.
  • a zosuquidar - cyclodextrin complex and a therapeutic agent that is a substrate for P-gp efflux can be administered to patients suffering from AML prior to confirmation of P-gp expression or function, or to AML patients other than relapse AML patients.
  • therapy is preferably administered to relapsed AML patients.
  • the administration route, amount administered, and frequency of administration can vary depending on the age of the patient, status as relapsed or newly diagnosed AML patient, and severity of the condition.
  • Contemplated amounts of Mylotarg for intravenous administration to treat relapsed AML are from about 10 mg/day or less to about 1000 mg/day or more administered on one, two, or more separate days.
  • the dosage is preferably administered intravenously at a rate of about 1 mg/m 2 or less to about 10 mg/m 2 or more continuously over the course of about 2, 3, or 4 hours to about 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, or 24 hours, more preferably over the course of about 2 hours to about 6 hours; however, administration at a rate of 5 mg/m 2 , 7 mg/m 2 , or 9 mg/m 2 over about 2 hours is particularly preferred.
  • doses of Mylotarg are administered on Day 1 and Day 15 of the treatment regimen.
  • the second dose can be administered on Day 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 16, 17, 18, 19, 20, 21, or 22, or another day of the treatment regimen.
  • Other dosing regimens include administering three doses total over a week.
  • Contemplated amounts of zosuquidar (in the form of a cyclodextrin complex) for intravenous administration to treat relapsed AML are from about 400 mg/day or less to about 1,600 mg/day or more, preferably from about 500, 600, or 700 mg/day to about 900, 1000, 1100, 1200, 1300, 1400, or 1500 mg/day, and most preferably from about 500 mg/day to about 800 mg/day.
  • the zosuquidar - cyclodextrin complex is preferably administered on two, three, or four separate days.
  • the dosage is preferably administered intravenously continuously over the course of about 6 to 90 hours, more preferably over the course of 12, 18, 24, 30, 36, or 42 hours to about 54, 60, 66, 72, 78, or 84 hours, most preferably over about 24 hours, 48 hours, or 72 hours, depending upon the treatment regimen.
  • the zosuquidar - cyclodextrin complex is administered on Day 1 of the treatment regimen.
  • additional zosuquidar - cyclodextrin complex is administered on Day 2, on Days 2 and 3, or on Days 2, 15, and 16.
  • one, two, or three or more additional doses can be administered on other days of the treatment regimen.
  • Table 1 provides various dosing regimes that can be used in treating relapsed AML.
  • Tables 2 and 3 provide alternative dosing regimes that can be used in treating relapsed AML.
  • a diagnostic or assay to determine P-gp expression or function or efflux pump activity can be useful in devising treatment regimens for other cancers, such as metastatic breast cancer, that also exhibit P-gp expression.
  • a P-gp expression or efflux pump activity diagnostic is conducted to provide information in treating newly diagnosed AML patients with a zosuquidar - cyclodextrin complex (e.g., zosuquidar - sulfobutylcyclodextrin or zosuquidar - hydroxypropyl cyclodextrin) in combination with daunombicin and cytarabine.
  • a zosuquidar - cyclodextrin complex e.g., zosuquidar - sulfobutylcyclodextrin or zosuquidar - hydroxypropyl cyclodextrin
  • P-gp expression or efflux pump activity diagnostic indicate negative P-gp expression
  • treatment with a P-gp efflux inhibitor is discontinued because administration of the drug is not expected to contribute to an improved clinical outcome.
  • P-gp expression or function or efflux pump activity is determined both in the presence and the absence the P-gp efflux inhibitor to determine the P-gp expression that is inhibitable by the P-gp efflux inhibitor.
  • Daunorubicin is an antibiotic chemotherapy treatment that is widely used to treat acute myeloid leukemia and acute lymphocytic leukemia. It is produced by the bacteria Streptomyces coeruleorubidis and was approved by the FDA as a first line therapy treatment for leukemia in 1998. Daunorubicin is typically administered intravenously. It is marketed under the brand names Cerubidine, DaunoXome, and Liposomal daunorubicin. Daunorubicin has the following structure:
  • Cytarabine is a deoxycytidine analogue, cytosine arabinoside (ara-C), which is metabolically activated to the triphosphate nucleotide (ara-CTP), which acts as a competitive inhibitor of DNA polymerase and produces S phase— specific cytotoxicity. It is used as an antineoplastic, generally as part of a combination chemotherapy regimen, in the treatment of acute lymphocytic and acute myelogenous leukemia, the blast phase of chronic myelogenous leukemia, erythroleukemia, and non-Hodgkin's lymphoma. It is typically administered intravenously and subcutaneously, and for the prophylaxis and treatment of meningeal leukemia, administered intrathecally. Cytarabine has the following structure:
  • the combination of a zosuquidar — cyclodextrin complex, the antibiotic chemotherapeutic daunorubicin, and the antineoplastic cytarabine is effective for treatment of newly diagnosed AML.
  • the effective dose of the zosuquidar - cyclodextrin complex and the timing of administration of the zosuquidar - cyclodextrin complex, daunorubicin, and cytarabine are critical to achieving improved complete remission rates and enhanced leukemia free survival rates in the newly diagnosed AML patient population. While the methods and formulations of preferred embodiments are especially preferred for treatment of newly diagnosed AML patients, the methods and formulations can be adapted to other drugs and indications.
  • chemotherapeutics other than daunorubicin and cytarabine can be administered according to the disclosed dosing regimens, or slightly modified dosing regimens.
  • the formulations and dosing regimens employing a zosuquidar - cyclodextrin complex, daunorubicin, and cytarabine can be employed in treating AML patients other than newly diagnosed AML patients, or for treating other types of leukemia or other cancers that exhibit P-gp expression.
  • Zosuquidar - cyclodextrin complex, daunorubicin, and cytarabine can be formulated as described above for zosuquidar - cyclodextrin complex and Mylotarg, and can be included in kits, also as described! a bove.
  • the zosuquidar - cyclodextrin complex, daunorubicin, and/or cytarabine can be administered to patients suffering from AML prior to confirmation of the P-gp expression or function, or to AML patients other than newly diagnosed AML patients (e.g., relapsed AML patients). However, therapy is preferably administered to newly diagnosed AML patients.
  • the administration route, amount administered, and frequency of administration can vary depending on the age of the patient, status as relapsed or newly diagnosed AML patient, and severity of the condition.
  • Contemplated amounts of zosuquidar (in the form of a cyclodextrin complex) for intravenous administration to treat newly diagnosed AML are from about 400 mg/day or less to about 1,600 mg/day or more, preierably trom aoout z> ⁇ , o ⁇ , or mg/ ⁇ ay io about 900, 1000, 1100, 1200, 1300, 1400, or 1500 mg/day, and most preferably 700 mg/day.
  • the zosuquidar - cyclodextrin complex is preferably administered on two, three, or four separate days.
  • the dosage is preferably administered in intravenously continuously over the course of about 6 to about 90 hours, more preferably over the course of about 12, 18, 24, 30, 36, or 42 hours to about 54, 60, 66, 72, 78, or 84 hours, most preferably over about 24 hours, 48 hours, or 72 hours, depending upon the treatment regimen.
  • the zosuquidar — cyclodextrin complex is administered on Day 1 of the treatment regimen.
  • additional zosuquidar - cyclodextrin complex is administered on Day 2, on Days 2 and 3, or on Days 2, 15, and 16.
  • one, two, or three or more additional doses can be administered on other days of the treatment regimen.
  • Contemplated amounts of daunorubicin for intravenous administration to treat newly diagnosed AML are from about 10 mg/m 2 /day or less to about 100 mg/m 2 /day or more administered at initiation of zosuquidar — cyclodextrin complex infusion or up to about 1, 2, 3, 4, 5, or 6 or more hours after initiation of zosuquidar — cyclodextrin complex infusion.
  • the dosage is preferably administered intravenously at a rate of about 25 mg/m 2 /day or less to about 90 mg/m 2 /day or more, preferably about 30, 35, or 40 mg/m 2 /day or less to about 50, 55, 60, 65, 70, 75, 80, or 85 mg/m 2 /day, and most preferably about 45 mg/m 2 /day continuously over the course of about 2 or 2.5 days to about 3.5 or 4 days, preferably about 3 days.
  • Contemplated amounts of cytarabine for intravenous administration to treat newly diagnosed AML patients are from about 10 mg/day or less to about 3,000 mg/day or more administered at initiation of zosuquidar - cyclodextrin complex infusion or after initiation of zosuquidar - cyclodextrin complex infusion.
  • the dosage is preferably administered intravenously at a rate of about 50 mg/m 2 /day or less to about 200 mg/m 2 /day or more, preferably 60, 70, 80, or 90 mg/m 2 /day or less to about 110, 120, 130, 140, 150, 160, 170, 180, or 190 mg/m 2 /day, and most preferably about 100 mg/m 2 /day continuously over the course of about 1, 2, 3, 4, 5, or 6 days up to about 8, 9, or 10 days or more, preferably over about 7 days.
  • a particularly preferred dosing regimen for newly diagnosed AML includes continuous intravenous administration of 550 mg of zosuquidar (as a cyclodextrin complex) over 6 hours (3 days), continuous intravenous administration of cytarabine at a rate of 100 mg/m 2 /day (7 days), and intravenous administration of daunorubicin at a dose of 45 mg/m 2 /day (3 days), wherein infusion of daunorubicin is started 1 hour after initiation of zosuquidar infusion.
  • Another particularly preferred dosing regimen includes continuous intravenous administration (preferably about 1 to 24 hours in duration, more preferably about 6 to 24 hours in duration, most preferably about 24 hours in duration) of 500 to 700 mg/day of zosuquidar (3 days), continuous intravenous administration of cytarabine at a rate of 100 mg/m 2 /day (7 days), and intravenous administration of daunorubicin at a dose of 45 mg/m 2 /day (3 days), wherein infusion of daunorubicin is started 1 to 4 hours after initiation of zosuquidar - cyclodextrin complex infusion.
  • infusion of daunorubicin is started after a specified time period has lapsed after initiation of zosuquidar - cyclodextrin complex infusion
  • other start times can be preferred, e.g., immediately after or during initiation of zosuquidar - cyclodextrin complex infusion up to about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, or more hours after initiation of zosuquidar - cyclodextrin complex infusion.
  • CAPTISOL® sulfobutylcyclodextrin
  • ⁇ -cyclodextrin derivative sodium salt CyDex, Inc., Lenexa, KS
  • CAPTISOL® is a polyanionic ⁇ -cyclodextrin derivative with a sodium sulfonate salt separated from the lipophilic cavity by a butyl ether spacer group, or sulfobutylether.
  • CAPTISOL® Upon intravenous administration, CAPTISOL® exhibits limited plasma protein binding and distributes to intracellular fluid. IV doses of 14 C-labeled CAPTISOL administered to rats, mice, dogs, rabbits and humans were rapidly and completely cleared intact from the circulation. Excretion is primarily in urine, with clearance approximating the glomerular filtration rate.
  • acceptable fill volumes for 800 mg zosuquidar can be achieved for solutions containing from about 8 mg/mL to about 50 mg/mL zosuquidar, and from about 5 wt. % (based on solvent, i.e., water, weight) to about 30 wt. % (based on solvent, i.e., water, weight) of sulfobutylcyclodextrin. Larger or smaller amounts of zosuquidar can be filled into vials by varying fill volume.
  • the data demonstrate that the solubility of zosuquidar is significantly increased when CAPTISOL® ( ⁇ -cyclodextrin derivative sodium salt) is incorporated into the formulation.
  • the graph in Figure 1 illustrates the increase of zosuquidar solution concentration as a function of sulfobutylcyclodextrin concentration.
  • Samples were formulated containing 20% CAPTISOL®, 33.3 mg/mL of zosuquidar, and different amounts of glycine. 2 mL aliquots were placed into 5 mL x 13 mm vials and were freeze-dried using a conservative cycle. Half of the samples were held aside and freeze-dried using a conservative cycle with an annealing step (hold at -15 0 C for 2 hours prior to re-cooling back to -45°C and freeze-drying). All of the vials contained yellow cakes, which were slightly shrunken, and no signs of collapse were observed. Samples were reconstituted with 2 mL of purified water, and the dissolution time and the description of the solution was recorded. Table 10 contains the samples tested and the results. Table 10.
  • Samples were prepared according to the concentrations listed in Table 12. 16 mL aliquots were filled into 30 mL x 20 mm tubing vials and lyophilized using a conservative cycle. The 16 mL fill samples were reconstituted with 20 mL of purified water, and the 8 mL fill samples were reconstituted with 10 mL of purified water.
  • the optimal concentration of CAPTISOL® and zosuquidar was 150 mg/mL (15%) and 22.5 mg/mL, respectively.
  • a drug product comprising 275 mg of zosuquidar trihydrochloride in CAPTISOL was formulated that exhibited superior solubility characteristics.
  • a fill volume of 12.2 mL per 30 mL vial was employed.
  • the total CAPTISOL® concentration per vial was 1.83 g. This concentration of CAPTISOL® solubilized zosuquidar and provided an acceptable reconstitution rate for the vial.
  • CAPTISOL® in combination with zosuquidar provides a stable formulation, as demonstrated by the real time stability data in Table 15. Table 15.

Abstract

La présente invention concerne un procédé de traitement de patients souffrant de leucémies, de tumeurs solides et autres malignités à l'aide d'agents chimiothérapeutiques en association avec du zosuquidar qui a été solubilisé par une cyclodextrine modifiée, telle qu'une sulfobutylcyclodextrine ou une hydroxypropyle cyclodextrine. L'invention concerne également des formulations pharmaceutiques comportant du zosuquidar en association avec une cyclodextrine modifiée.
PCT/US2006/026031 2005-07-06 2006-06-30 Formulations chimiotherapeutiques de trihydrochlorure de zosuquidar et cyclodextrines modifies WO2007008496A1 (fr)

Priority Applications (3)

Application Number Priority Date Filing Date Title
EP06774486A EP1898957A1 (fr) 2005-07-06 2006-06-30 Formulations chimiothérapeutiques de trihydrochlorure de zosuquidar et cyclodextrines modifiés
AU2006269498A AU2006269498A1 (en) 2005-07-06 2006-06-30 Chemotherapeutic formulations of zosuquidar trihydrochloride and modified cyclodextrins
CA002630087A CA2630087A1 (fr) 2005-07-06 2006-06-30 Formulations chimiotherapeutiques de trihydrochlorure de zosuquidar et cyclodextrines modifies

Applications Claiming Priority (22)

Application Number Priority Date Filing Date Title
US69675605P 2005-07-06 2005-07-06
US69693005P 2005-07-06 2005-07-06
US69693905P 2005-07-06 2005-07-06
US60/696,930 2005-07-06
US60/696,756 2005-07-06
US60/696,939 2005-07-06
US11/416,829 US20070010478A1 (en) 2005-07-06 2006-05-03 Zosuquidar, daunorubicin, and cytarabine for the treatment of cancer
US11/417,958 US20070010485A1 (en) 2005-07-06 2006-05-03 Chemotherapeutic formulations of zosuquidar trihydrochloride and modified cyclodextrins
US11/417,958 2006-05-03
US11/418,400 2006-05-03
US11/416,571 US20070010465A1 (en) 2005-07-06 2006-05-03 Zosuquidar, daunorubicin, and cytarabine for the treatment of cancer
US11/416,829 2006-05-03
US11/418,324 2006-05-03
US11/416,832 2006-05-03
US11/416,992 2006-05-03
US11/416,992 US20070009532A1 (en) 2005-07-06 2006-05-03 Treatment of patients with cancer using a calicheamicin-antibody conjugate in combination with zosuquidar
US11/418,324 US20070010486A1 (en) 2005-07-06 2006-05-03 Chemotherapeutic formulations of zosuquidar trihydrochloride and modified cyclodextrins
US11/416,833 2006-05-03
US11/416,571 2006-05-03
US11/416,833 US20070009531A1 (en) 2005-07-06 2006-05-03 Treatment of patients with cancer using a calicheamicin-antibody conjugate in combination with zosuquidar
US11/418,400 US20070010487A1 (en) 2005-07-06 2006-05-03 Chemotherapeutic formulations of zosuquidar trihydrochloride and modified cyclodextrins
US11/416,832 US20070010466A1 (en) 2005-07-06 2006-05-03 Zosuquidar, daunorubicin, and cytarabine for the treatment of cancer

Publications (1)

Publication Number Publication Date
WO2007008496A1 true WO2007008496A1 (fr) 2007-01-18

Family

ID=37637468

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2006/026031 WO2007008496A1 (fr) 2005-07-06 2006-06-30 Formulations chimiotherapeutiques de trihydrochlorure de zosuquidar et cyclodextrines modifies

Country Status (4)

Country Link
EP (1) EP1898957A1 (fr)
AU (1) AU2006269498A1 (fr)
CA (1) CA2630087A1 (fr)
WO (1) WO2007008496A1 (fr)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2132173A1 (fr) * 2007-02-26 2009-12-16 Merck & Co., Inc. Formulations pour inhibiteurs de la cathepsine k
CN103142499A (zh) * 2013-03-21 2013-06-12 青岛正大海尔制药有限公司 一种依托泊苷颗粒
CN113891725A (zh) * 2019-10-31 2022-01-04 南京海维医药科技有限公司 包含血管紧张素ii的固态组合物及其制备方法、使用方法、用途

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5134127A (en) * 1990-01-23 1992-07-28 University Of Kansas Derivatives of cyclodextrins exhibiting enhanced aqueous solubility and the use thereof
US5643909A (en) * 1993-04-19 1997-07-01 Syntex (U.S.A.) Inc. 10,11-Methanodibenzosuberane derivatives

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5134127A (en) * 1990-01-23 1992-07-28 University Of Kansas Derivatives of cyclodextrins exhibiting enhanced aqueous solubility and the use thereof
US5643909A (en) * 1993-04-19 1997-07-01 Syntex (U.S.A.) Inc. 10,11-Methanodibenzosuberane derivatives

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2132173A1 (fr) * 2007-02-26 2009-12-16 Merck & Co., Inc. Formulations pour inhibiteurs de la cathepsine k
EP2132173A4 (fr) * 2007-02-26 2010-06-02 Merck Sharp & Dohme Formulations pour inhibiteurs de la cathepsine k
CN103142499A (zh) * 2013-03-21 2013-06-12 青岛正大海尔制药有限公司 一种依托泊苷颗粒
CN113891725A (zh) * 2019-10-31 2022-01-04 南京海维医药科技有限公司 包含血管紧张素ii的固态组合物及其制备方法、使用方法、用途

Also Published As

Publication number Publication date
AU2006269498A1 (en) 2007-01-18
EP1898957A1 (fr) 2008-03-19
CA2630087A1 (fr) 2007-01-18

Similar Documents

Publication Publication Date Title
CA2763365C (fr) Compositions injectables a base de melphalan comprenant un derive de cyclodextrine et leurs procedes de fabrication et d'utilisation
CN103705942B (zh) 抗微生物组合物
CN1612737A (zh) 铂衍生物药物制剂
CN1623543A (zh) 依普西龙组合物
CN108136217B (zh) 用于治疗膀胱癌的制剂
US20070010466A1 (en) Zosuquidar, daunorubicin, and cytarabine for the treatment of cancer
AU2019373373B2 (en) Cyclodextrin-based formulation of a Bcl-2 inhibitor
US20070009532A1 (en) Treatment of patients with cancer using a calicheamicin-antibody conjugate in combination with zosuquidar
WO2007008496A1 (fr) Formulations chimiotherapeutiques de trihydrochlorure de zosuquidar et cyclodextrines modifies
JP2011529950A (ja) 化学療法剤の経口製剤
US20070010486A1 (en) Chemotherapeutic formulations of zosuquidar trihydrochloride and modified cyclodextrins
CN101137364B (zh) 包含埃坡霉素的组合物及其制备方法
US20070010485A1 (en) Chemotherapeutic formulations of zosuquidar trihydrochloride and modified cyclodextrins
JP5466174B2 (ja) 水溶性、カチオン性および両親媒性の薬学的に活性な物質を投与するためのドラッグ・デリバリー・システム
EP2934593B1 (fr) Composition de cabazitaxel
US20070010487A1 (en) Chemotherapeutic formulations of zosuquidar trihydrochloride and modified cyclodextrins
CN106902357B (zh) 药物组合物及其应用、药物包合物、静脉制剂及制备方法
EP1898916A2 (fr) Zosuquidar, daunorubicine et cytarabine pour le traitement du cancer
WO2013026694A1 (fr) Compositions pharmaceutiques comprenant du voriconazole
JP6654702B2 (ja) 経口製剤およびその製造方法
US20240016948A1 (en) Single protein-encapsulated pharmaceutics for enhancing therapeutic effects
RU2804366C2 (ru) Состав ингибитора bcl-2 на основе циклодекстрина
CN117580803A (zh) 用于药物递送的硅颗粒
EA044714B1 (ru) Состав ингибитора bcl-2 на основе циклодекстрина
Capasso SVILUPPO, CARATTERIZZAZIONE E VALUTAZIONEDI CARRIER COLLOIDALI O MICROPARTICELLARIPER LA VEICOLAZIONE DI FARMACI

Legal Events

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

Ref document number: 2630087

Country of ref document: CA

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2006774486

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2006269498

Country of ref document: AU

ENP Entry into the national phase

Ref document number: 2006269498

Country of ref document: AU

Date of ref document: 20060630

Kind code of ref document: A