WO2009089366A2 - Suppression of cancer growth and metastasis using nordihydroguaiaretic acid derivatives with metabolic modulators - Google Patents

Suppression of cancer growth and metastasis using nordihydroguaiaretic acid derivatives with metabolic modulators Download PDF

Info

Publication number
WO2009089366A2
WO2009089366A2 PCT/US2009/030459 US2009030459W WO2009089366A2 WO 2009089366 A2 WO2009089366 A2 WO 2009089366A2 US 2009030459 W US2009030459 W US 2009030459W WO 2009089366 A2 WO2009089366 A2 WO 2009089366A2
Authority
WO
WIPO (PCT)
Prior art keywords
tumor
inhibitor
cells
amino acid
acid residue
Prior art date
Application number
PCT/US2009/030459
Other languages
French (fr)
Other versions
WO2009089366A3 (en
Inventor
Ru Chih C. Huang
Kotohiko Kimura
Original Assignee
The Johns Hopkins University
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
Application filed by The Johns Hopkins University filed Critical The Johns Hopkins University
Priority to US15/725,624 priority Critical patent/USRE46907E1/en
Priority to US12/812,196 priority patent/US9149526B2/en
Priority to EP09701300.7A priority patent/EP2240168B1/en
Publication of WO2009089366A2 publication Critical patent/WO2009089366A2/en
Publication of WO2009089366A3 publication Critical patent/WO2009089366A3/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/02Halogenated hydrocarbons
    • A61K31/025Halogenated hydrocarbons carbocyclic
    • A61K31/03Halogenated hydrocarbons carbocyclic aromatic
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents

Definitions

  • This invention relates to the use of nordihydroguaiaretic acid derivatives together with one or more metabolic modulators to treat cancer, prevent metastasis, and prolong the life of a mammal afflicted with a tumor.
  • Carcinogenesis is a multistage event affected by a variety of genetic and epig ⁇ n ⁇ tic factors and is typified by the outbreak of uncontrolled cell growth originated from different tissues.
  • a universal goal for anticancer research lies in the development of a clinical treatment that is highly effective in curtailment of tumor growth, non-toxic to the host, and is affordable for most patients.
  • Drugs that inhibit targets that are unique to dividing cells, particularly cells dividing in an uncontrolled manner are an ideal paradigm for chernotherapeutic agents, the greater the specificity to cells that are dividing in an uncontrolled manner the lower the risk of attendant side effects.
  • Survivin is one of the targets currently being tested for anticancer therapy,
  • Cdc2 cyclin-dependent kinase-1
  • Cdc2 is one of several kinase proteins controlling cell division and is frequently de-regulated in cancer cells.
  • Cdc2 is also involved in the activation of survivin.
  • growing tumors require a constant supply of essential nutrients and oxygen.
  • Cancer cells achieve their nutrient needs by secreting Vascular Endothelial Growth Factor (VEGF) to induce new blood vessel formation within the tumor mass.
  • VEGF Vascular Endothelial Growth Factor
  • Proteins such as those mentioned above are involved in the regulation of the signaling pathways that control cell growth, division and death. Some of these proteins are significantly altered in cancer cells. The process of converting the gene sequence on DNA to an RNA message (a process called transcription) that can then be converted to protein (a process called translation) is essential to the regulation of protein production.
  • Cells pass through many checkpoints as they proceed through the eel! cycle. Certain criteria must be met in order to pass each of these checkpoints. In the G2/M transition, the most essential regulator is the cyclin-dependent kinase CDC2.
  • This kinase binds tightly to the regulatory protein cyclin B, and this complex, also called the maturation promoting factor (MPF), is responsible for stimulating a myriad of events that lead to the cell's entry into early prophase (1), Not surprisingly, the loss or deactivation of either component of the MPF will block cellular progression out of G2.
  • MPF maturation promoting factor
  • Survivin is an inhibitor of apoptosis that is abundantly expressed in many human cancers (7), but not in normal adult human tissue, and is considered a possible modulator of the terminal effector phase of cell death/survival.
  • Survivin is expressed in G 2 -M in a cell cycle-dependent manner, binding directly to mitotic spindle microtubules.
  • NDGA Jignan nordihydroguaiaretic acid
  • M 4 N 5 EM 1421, Terameprocol is the synthetic telra- methylated derivative of nordihydroguaiaretic acid (tetra-O-methyl nordihydroguaiaretic acid, abbreviated as M 4 N),
  • M 4 N tetra-O-methyl nordihydroguaiaretic acid
  • M 4 N The chemical structure of M 4 N was designed to make it pharmacologically distinct from NDGA.
  • M 4 N has been shown to possess antiviral (12, 14) and anti-cancer (16) activities in cultured cells, in mouse models (16, 17), and in human xenografts in nude mice (18).
  • M 4 N causes cell cycle arrest at the G2 phase of the cell cycle probably by suppressing Sp-I regulated cdk expression (16, 19).
  • M 4 N has been in Phase I clinical trials in patients by intravenous infusion (CLINICAL TRIALS.GOV, A service of U.S. NIH).
  • Nordihydroguaiaretic acid (NDGA) derivatives such as M 4 N suppress SpI regulated transcription of viral genes, by deactivation of SpI -dependent promoters. SPl also affects expression of many growth-related genes.
  • Cdc2 also referred to as CDKl
  • cyclin B interact to allow cells to move from the G2 phase of celi division to mitosis.
  • M 4 N blocks the transcription of Cdc2, and thus blocks cell division.
  • the Sp 1 protein on promoter of CDC2 chromatin is replaced following M 4 N treatment in vivo.
  • M 4 N is able to induce cell cycle arrest in mammalian cell lines; M 4 N is a transcription inhibitor. It selectively reduces transcription of growth related genes that have promoters controlled by the SpI factor, such as cdc2, survivin and VEGF. By blocking production of cdc2, and VEGF, M 4 N inhibits tumor growth and starves nmiors by restricting growth of their blood supply.
  • M J N has been shown to arrest growth of a variety of human cells in vitro, the majority of which are part of the NCI panel of 60 cancer cell lines, including solid tumor cell lines (bladder, breast, colorectal, liver, lung, ovarian, pancreatic, prostate and cervical carcinomas), and erythroleukemia cells.
  • solid tumor cell lines bladedder, breast, colorectal, liver, lung, ovarian, pancreatic, prostate and cervical carcinomas
  • erythroleukemia cells In vivo, M 4 N also decreases tumor cell growth and exhibits antitumor activity in a large number of tumor xenograft models, including human bladder, breast, colorectal, liver, ovarian, pancreatic, prostate and cervical carcinomas, and erythroleukemia, without apparent toxicity.
  • MjN has a broad spectrum of activity in anticancer therapy, having affects on Cdc2, HIF- l ⁇ , MDRl, VEGF and survivin.
  • M 4 N induces apopiosis and reduces cdc2 protein levels in human oral cancers. M 4 N also appears to reduce survivin levels in these cancers.
  • Administered systemically, M 4 N was also shown to inhibit xenografted human tumors MCF-7, Hep3b, LNCaP, HT-29, and Iv562, Although none of the xenograft tumors were fully eradicated,
  • M 4 N does not appear toxic to animals.
  • M 4 N retention in mouse organs following oral administration has been studied after short term and long term feeding, the results showed essentially no toxic effects even at concentrations high as 906 ⁇ g/g of tissue.
  • M 4 N On daily (1 rng/day) IV injection of M 4 N for days, M 4 N accumulated in blood and tumors to levels above 1 mM in nude mice carrying human tumor xenografts.
  • M 4 N has some favorable therapeutic qualities, in that it exhibits efficacy against several tumors, by inhibiting cell growth.
  • treatment with M 4 N does not generally eradicate disease, and upon cessation of treatment with M 4 N tumors are capable of growing back. Accordingly, there is a need to identify ways to boost the efficacy of the M 4 N when the type of cancer is aggressive, metastatic or when M 4 N as a single drug in low concentration is not enough to induce rapid apoptosis of such type of cancers.
  • Ri, R 2 , R 3 and R 4 independently represent hydroxy, a straight or branched chain lower alkyl or alkoxy, an amino acid residue, a substituted amino acid residue, a nitrogen- containing 5- or 6-membered heterocyclic ring or a saccharide residue; the amino acid residue, substituted amino acid residue, nitrogen-containing 5 or 6 membered heterocyclic ring or saccharide residue being optionally joined to the phenyl ring by a linker of an oxygen atom and 1-10 carbon atoms, and an effective amount of a metabolic modulator.
  • the pharmaceutical composition comprises the NDGA derivative tetra- ⁇ methyl nordihydroguaiaretic acid (M 4 N).
  • the pharmaceutical composition comprises the NDGA derivative maltose M 3 N (90).
  • Rj, R 2 , R 3 and R 4 are identical and represent straight or branched chain lower alkoxy groups or naturally occurring amino acid residues.
  • the metabolic modulator can be selected, for example from the group consisting of an inhibitor of the PDK-1/PI3K/AKT pathway, an inhibitor of the PKC ⁇ /topoisomerase ⁇ pathway, an inhibitor of mitochondrial permeability transition, and an inhibitor of the mitogen-activated protein kinase (MAPK/RAS) pathway, or any other metabolic modulators which can systematically induce translocation of phosphory ⁇ ated PC& ⁇ to the nucleus of tumor cells.
  • Examples of such metabolic inhibitors are Ly294002, etoposide, Rottlerin, dichloroacetate, c ⁇ tuximab, trastuzurnab, bevacizumab and rapamyein.
  • the metabolic inhibitor is etoposide, rapamycin, rottlerin and/or water soluble derivatives thereof, such as temsirolimus, everolimus and other selected molecular targeting agents in clinical development (Nature Reviews Drug Discovery vol 5, p 650, 2006).
  • a method of treating a tumor comprising administering to a mammal in need of treatment an effective amount of the above-described pharmaceutical composition.
  • the mammal can be any mammal afflicted with a tumor amenable to treatment, for example a human, nonhuman primate, cat, dog, mouse, etc.
  • the tumor may be a solid or hematological tumor, benign or malignant (metastatic or nonm ⁇ tastatic), such as, for example, breast, liver, prostate, cervical, ovarian, colon, brain, pancreatic, bladder esophagus, gut, head and neck, kidney, melanoma, stomach, testes, thyroid, uterine and lung cancers, leukemias and lymphomas, such as acute myelogenous leukemia, acute or chronic lymphocytic leukemia, Hodgkin's and non-Hodgkin lymphoma, and myelomas.
  • the treatment method is particularly suitable for treatment of metastatic and nonmetastatic cancer.
  • NDGA nordihydroguaiaretic acid
  • Rj 5 R 2 , R 3 and R 4 independently represent hydroxy, a straight or branched chain lower alkyl or alkoxy, an amino acid residue, a substituted amino acid residue, a nitrogen- containing 5- or 6 ⁇ membered heterocyclic ring or a saccharide residue; the amino acid residue, substituted amino acid residue, nitrogen-containing 5 or 6 membered heterocyclic ring or saccharide residue being optionally joined to the phenyl ring by a linker of an oxygen atom and 1-10 carbon atoms, and an effective amount of a metabolic modulator.
  • the NDGA derivative is tetra-o-methyl nordihydroguaiaretic acid (M 4 N).
  • the N DGA derivative is maltose M3N (90).
  • R 1 , R 2 , R 3 and R 4 are identical and represent straight or branched chain lower alkoxy groups or naturally occurring amino acid residues.
  • the mammal can be any mammal afflicted with a tumor amenable to treatment, for example a human, nonhuman primate, cat, dog, mouse, etc.
  • the metabolic inhibitor is selected from the group consisting of an inhibitor of the PDK-1/PI3K/AKT pathway, an inhibitor of the PKC ⁇ /topoisomerase Il ⁇ pathway, an inhibitor of mitochondrial permeability transition, and an inhibitor of the mitogen-activated protein kinase (MAPK/RAS) pathway.
  • the metabolic inhibitor is selected from the group consisting of Ly294002, etoposide, Rottlerin, dichloroacetate, cetuximab, trastuzumab, bevacizumab and rapamycin. Etoposide, rapamycin, and water soluble derivatives thereof are expected to be particularly effective.
  • the tumor may be a solid or hematological tumor, benign or malignant (metastatic or nonmetastatic), such as, for example, breast, prostate, cervical, ovarian, colon, brain, pancreatic and lung cancers, leukemias and lymphomas, and others mentioned hereinabove.
  • metalastatic or nonmetastatic such as, for example, breast, prostate, cervical, ovarian, colon, brain, pancreatic and lung cancers, leukemias and lymphomas, and others mentioned hereinabove.
  • the treatment method is particularly suitable for treatment of metastatic and nonmetastatic cancer.
  • NDGA nordihydroguaiaretic acid
  • R;, R 2 , R. 3 and R 4 independently represent hydroxy, a straight or branched chain lower aikyl or alkoxy s an amino acid residue, a substituted amino acid residue and a saccharide residue; the amino acid residue, substituted amino acid residue or saccharide residue being optionally joined to the phenyl ring by a Maker of an oxygen atom and 1-10 carbon atoms; and an effective amount of a metabolic modulator.
  • the NDGA derivative is ietra ⁇ o ⁇ methyl nordihydroguaiaretic acid (M 4 N), In another specific embodiment, the NDCJA derivative is maltose M 3 N (90).
  • Rj, R:,, R 3 and R 4 are identical and represent straight or branched chain lower alkoxy groups or naturally occurring amino acid residues.
  • the mammal can be any mammal afflicted with a tumor amenable to treatment, for example a human, nonhuman primate, cat, dog, mouse, etc.
  • the metabolic inhibitor is selected from the group consisting of an inhibitor of the PDK-1/PI3K/AKT pathway, an inhibitor of the PKC ⁇ /topoisomerase Il ⁇ pathway, an inhibitor of mitochondrial permeability transition, and an inhibitor of the mitogen-activated protein kinase (MAPK/RAS) pathway.
  • the metabolic inhibitor is selected from the group consisting of Ly294002, efoposide, Rottl ⁇ rin, dichloroacetate, cetuxtmab, trastuzu ⁇ iab, bevaeizumab and rapamycin.
  • Etoposide, rapamycin, and water soluble derivatives thereof are expected to be particularly effective.
  • the tumor may be a solid or hematological tumor, such as, for example, breast, prostate, cervical, ovarian, colon, brain, pancreatic and lung cancers, leukemias and lymphomas, and others mentioned hereinabove, Persons of skill in the art will be able to determine by routine experimentation the types of tumors that are amenable to treatment.
  • a solid or hematological tumor such as, for example, breast, prostate, cervical, ovarian, colon, brain, pancreatic and lung cancers, leukemias and lymphomas, and others mentioned hereinabove.
  • Treatment may be administered alone, or as an adjuvant to surgery, e.g. before surgery, for example, to reduce tumor size, and/or following surgery to reduce the possibility of metastases, e.g. by inhibition of the growth and migration of circulating tumor cells through the blood stream,
  • the invention also provides a method of prolonging the life of a mammal having a malignant tumor, said method comprising administering to the mammal an effective amount of nordihydroguaiaretic acid (NDGA) or a derivative thereof of formula I:
  • NDGA nordihydroguaiaretic acid
  • R-., R,J, R 3 and R4 independently represent hydroxy, a straight or branched chain lower alkyS or alkoxy, an amino acid residue, a substituted amino acid residue, and a saccharide residue; the amino acid residue, substituted amino acid residue or saccharide residue being optionally joined to the phenyl ring by a linker of an oxygen atom and 1-10 carbon atoms; and an effective amount of a metabolic modulator.
  • the NDGA derivative is tetra-o-methyl nofdihydroguaiaretic acid (M 4 N), In another specific embodiment, the NDGA derivative is maltose M3N (90). In other specific embodiments, Ri, R 2 , R 3 and R4 are identical and represent straight or branched chain lower alkoxy groups or naturally occurring amino acid residues.
  • the mammal can be any mammal afflicted with a tumor amenable to treatment, for example a human, nonhuman primate, cat, dog, mouse, etc.
  • the metabolic inhibitor is selected from the group consisting of an inhibitor of the PDK-I /PB K/ AKT pathway, an inhibitor of the PKC ⁇ /topoisomerase ⁇ pathway, an inhibitor of mitochondrial permeability transition, and an inhibitor of the mitogen-activated protein kinase (MAPIC 7 RAS) pathway.
  • the metabolic inhibitor is selected from the group consisting of Ly294002, etoposide, Rottlerin, dichloroacetate, cetuximab, trastuzumab, bevacizumab and rapamycin.
  • Etoposide, rapamycin, and water soluble derivatives thereof are expected to be particularly effective.
  • the tumor may be a solid or hematological tumor, such as, for example, breast, prostate, cervical, ovarian, colon, brain, pancreatic and lung cancers, leukemias and lymphomas, and other tumors mentioned hereinabove. Persons of skill in the art will be able to determine by routine experimentation the types of tumors that are amenable to treatment.
  • compositions in accordance with the invention are useful for diagnosis, prognosis , prophylaxis or treatment of a condition. Accordingly, compositions in accordance with the invention are useful as a drug or as information for structural modification of existing compounds, e.g., by rational drug design. Compounds and methods of the invention are useful for screening compounds having an effect on a variety of conditions. [040] For therapeutic uses, the compositions or agents identified using the methods disclosed herein may be administered systemically, for example, formulated in a pharmaceutically-acceptable buffer such as physiological saline.
  • Preferable routes of administration include, for example, subcutaneous, intravenous, intraperitoneally., intramuscular, or intradermal injections that provide continuous, sustained levels of the drug in the patient, Treatment of human patients or other animals are generally carried out using a therapeutically effective amount of a therapeutic of the invention in a physiologically- acceptable carrier, Suitable carriers and their formulation are described, for example, in Remington's Pharmaceutical Sciences by E. W. Martin.
  • the amount of the therapeutic agent to be administered varies depending upon the manner of administration, the age and body weight of the subject/patient, and with the subject's symptoms and condition. A compound is administered at a dosage that best achieves medical goals with the fewest corresponding side effects. [042] Administration
  • compositions of this invention including biologically active fragments, variants, or analogs thereof, can be administered by any suitable routes including intracranial, intracerebral, intraventricular, intrathecal, intraspinal, oral, topical, rectal, transdermal, subcutaneous, intravenous, intramuscular, intranasal, and the like.
  • the compositions are added to a retained physiological fluid, such as cerebrospinal fluid, blood, or synovial fluid.
  • the compositions of the invention can be amenable to direct injection or infusion at a site of disease or injury.
  • compositions for parenteral use may be provided in unit dosage forms (e.g., in single-dose ampules), or in vials containing several doses and in which a suitable preservative may be added.
  • the composition may be in the form of a solution, a suspension, an emulsion, an infusion device, or a deliver ⁇ ' device for implantation, or it may be presented as a dry powder to be reconstituted with water or another suitable vehicle before use.
  • the composition may include suitable parenterally acceptable carriers and/or excipients.
  • a therapeutic of the invention is provided within an implant, such as an osmotic pump, or in a graft comprising appropriately transformed cells.
  • Methods of introduction may also be provided by rechargeable or biodegradable devices.
  • Various slow release polymeric devices have been developed and tested for the controlled delivery of drugs, including proteinacious biopharmaceuticals.
  • a variety of biocompatible polymers including hydrog ⁇ ls), including both biodegradable and non-degradable polymers, can be used to form an implant for the sustained release of a bioactive factor at a particular target site.
  • the amount of administered agent of the invention (dosage) will be empirically determined in accordance with information and protocols known in the art.
  • agents are administered in the range of about 10 ⁇ g/kg to 100 mg/kg of the recipient.
  • Other additives may be included, such as stabilizers, bactericides, and anti-fungals.
  • Columns 2 and 3 of Table 1 show effective dosages administered in mice in experiments as described herein, and administered to humans in clinical trials.
  • Column 4 shows Km factor for conversion to patient treatment dosage.
  • Column 5 gives the calculated amount for administration to patients. In general it is expected that blood levels of ,5 to 10 mM of M4N, preferably 1-5 mM, should be achieved for effective patient treatment. Persons of skill in the art will appreciate that the dosage is expected to vary depending on the protocol used for drug administration.
  • the administration of a compound of the invention may be by any suitable means that results in a concentration of the therapeutic that, combined with other components, is effective in ameliorating, reducing, or stabilizing a deficit or disorder.
  • the compound may be contained in any appropriate amount in any suitable carrier substance, and is generally present in an amount of 1-95% by weight of the total weight of the composition.
  • the composition may be provided in a dosage form that is suitable for parenteral (e.g., subcutaneously, intravenously, intramuscularly, or intraperitoneally) administration route.
  • the pharmaceutical compositions may be formulated according to conventional pharmaceutical practice (see, e.g., Remington: The Science and Practice of Pharmacy (20th ed.), ed. A. R. Gennaro, Lippincott Williams & Wilkins, 2000 and Encyclopedia of Pharmaceutical Technology, eds. J. Swarbrick and J. C. Boylan, 1988-1999, Marcel Dekker, New York).
  • compositions of the invention can be administered parenterally by injection, infusion or implantation (subcutaneous, intravenous, intramuscular, intraperitoneal, or the like) in dosage forms, formulations, or via suitable delivery devices or implants containing conventional, non-toxic pharmaceutically acceptable carriers and adjuvants.
  • injection, infusion or implantation subcutaneous, intravenous, intramuscular, intraperitoneal, or the like
  • suitable delivery devices or implants containing conventional, non-toxic pharmaceutically acceptable carriers and adjuvants.
  • formulation and preparation of such compositions are well known to those skilled in the art of pharmaceutical formulation. Formulations can be found in Remington: The Science and Practice of Pharmacy, cited herein.
  • compositions according to the invention may be in the form suitable for sterile injection.
  • the compositions(s) are dissolved or suspended in a parenterally acceptable liquid vehicle.
  • acceptable vehicles and solvents that may be employed are water, water adjusted to a suitable pH by addition of an appropriate amount of hydrochloric acid, sodium hydroxide or a suitable buffer, 1,3-butanediol, Ringer's solution, and isotonic sodium chloride solution and dextrose solution.
  • the aqueous formulation may also contain one or more preservatives (e.g., methyl, ethyl or n-propyl p-hydroxybenzoate).
  • Suitable dosage forms can be formulated for, but are not limited to, oral, rectal, sub-lingual, mucosal, nasal, ophthalmic, subcutaneous, intramuscular, intravenous, transdermal, spinal, intrathecal, intra-articular, intra-art ⁇ rial. sub-arachinoid, bronchial, lymphatic, and intra-uterille administration, and other dosage forms for systemic delivery of active ingredients.
  • the dosage form is suitable for injection or intravenous administration.
  • one or more of the aforementioned compounds are intimately admixed with a pharmaceutical carrier according to conventional pharmaceutical compounding techniques.
  • the carrier may take a wide variety of forms depending on the form of preparation desired for administration.
  • the carrier will usually comprise sterile water, though other ingredients, for example, ingredients that aid solubility or for preservation, may be included. Injectable solutions may also be prepared in which case appropriate stabilizing agents may be employed.
  • any of the usual pharmaceutical media may be employed.
  • suitable carriers and additives include water, glycols, oils, alcohols, flavoring agents, preservatives, coloring agents and the like.
  • suitable carriers and additives include starches, sugars, diluents, granulating agents, lubricants, binders, disintegrating agents and the like. Due to their ease in administration, tablets and capsules represent the most advantageous oral dosage unit form, [f desired, tablets may be sugar coated or enteric coated by standard techniques.
  • vectorized form such as by encapsulation of the active agent in a liposome or other encapsulant medium, or by fixation of the active agent, e.g., by covalent bonding, chelation, or associative coordination, on a suitable biomoleeule, such as those selected from proteins, lipoproteins, glycoproteins, and polysaccharides.
  • compositions suitable for oral administration may be presented as discrete units such as capsules, cachets, tablets, or lozenges, each containing a predetermined amount of the active ingredient as a powder or granules.
  • a suspension in an aqueous liquor or a non-aqu ⁇ ous liquid may be employed, such as a syrup, an elixir, an emulsion, or a draught.
  • Formulations for oral use include tablets containing active ingredient(s) of the invention in a mixture with pharmaceutically acceptable excipients, Such formulations are known to the skilled artisan.
  • Excipients may be, for example, inert diluents or fillers (e.g., sucrose, sorbitol, sugar, manniiol, mieroeryslall ⁇ ne cellulose, starches including potato starch.
  • granulating and disintegrating agents e.g., cellulose derivatives including microcrystalline cellulose, starches including potato starch, croscarmellose sodium, alginates, or alginic acid
  • binding agents e.g., sucrose, glucose, sorbitol, acacia, alginic acid, sodium alginate, gelatin, starch, prege latinized starch, microcrystalline cellulose, magnesium aluminum silicate, carboxymethylcellulose sodium, methylcellulose, hydroxypropyl methylcellulose, ethylcellulose, polyvinylpyrrolidone, or polyethylene glycol
  • lubricating agents, glidants, and antiadhesives e.g., magnesium stearat ⁇ , zinc st ⁇ arate, stearic acid, silicas, hydrogenated vegetable oils, or talc.
  • Other pharmaceutically acceptable excipients can be colorants,
  • a tablet may be made by compression or molding, or wet granulation, optionally with one or more accessory ingredients.
  • Compressed tablets may be prepared by compressing in a suitable machine, with the active compound being in a free- flowing form such as a powder or granules which optionally is mixed with a binder, disintegrant, lubricant, inert diluent, surface active agent, or discharging agent.
  • Molded tablets comprised of a mixture of the powdered active compound with a suitable carrier may be made by molding in a suitable machine.
  • a syrup may be made by adding the active compound to a concentrated aqueous solution of a sugar, for example sucrose, to which may also be added any accessory ingredient(s).
  • a sugar for example sucrose
  • Such accessory ingredient(s) may include flavorings, suitable preservative, agents to retard crystallization of the sugar, and agents to increase the solubility of any other ingredient, such as a polyhydroxy alcohol, for example glycerol or sorbitol.
  • Formulations suitable for parenteral administration usually comprise a sterile aqueous preparation of the active compound, which preferably is isotonic with the blood of the recipient (e.g., physiological saline solution).
  • Such formulations may include suspending agents and thickening agents and liposomes or other microparticulate systems which are designed to target the compound to blood components or one or more organs.
  • the formulations may be presented in unit-dose or multi-dose form.
  • Parenteral administration may comprise any suitable form of systemic delivery or localized delivery. Administration may for example be intravenous, intra-arterial, intrathecal, intramuscular, subcutaneous, intramuscular, intra-abdominal (e.g., intraperitoneal), etc., and may be effected by infusion pumps (external or implantable) or any other suitable means appropriate to the desired administration modality.
  • Nasal and other mucosal spray formulations e.g. inhalable forms
  • Such formulations are preferably adjusted to a pH and isotonic state compatible with the nasal or other mucous membranes.
  • formulations for rectal administration may be presented as a suppository with a suitable carrier such as cocoa butter, hydrogenated fats, or hydrogenated fatty carboxylic acids.
  • Transdermal formulations may be prepared by incorporating the active agent in a thixotropic or gelatinous carrier such as a cellulosic medium, e.g., methyl cellulose or hydroxyethyl cellulose, with the resulting formulation then being packed in a transdermal device adapted to be secured in dermal contact with the skin of a wearer.
  • a thixotropic or gelatinous carrier such as a cellulosic medium, e.g., methyl cellulose or hydroxyethyl cellulose
  • formulations of the invention may further include one or more accessory ingredient(s) selected from diluents, buffers, flavoring agents, binders, disintegrants, surface active agents, thickeners, lubricants, preservatives (including antioxidants), and the like.
  • a formulation of the present invention can have immediate release, sustained release, delay ⁇ d-onset release or any other release profile known to one skilled in the art.
  • Pharmaceutical compositions according to the invention may be formulated to release the active compound substantially immediately upon administration or at any predetermined time or time period after administration.
  • compositions are generally known as controlled release formulations, which include (i) formulations that create a substantially constant concentration of the drug within the bodv over an extended period of time; (ii) formulations that after a predetermined lag time create a substantially constant concentration of the drug within the body over an extended period of time; (iii) formulations that sustain action during a predetermined time period by maintaining a relatively, constant, effective level in the body with concomitant minimization of undesirable side effects associated with fluctuations in the plasma level of the active substance (sawtooth kinetic pattern); (iv) formulations that localize action by, e.g., spatial placement of a controlled release composition adjacent to or in the central nervous system or cerebrospinal fluid; (v) formulations that allow for convenient dosing, such that doses are administered, for example, once every one or two weeks; and (vi) formulations that target the site of a pathology.
  • controlled release formulations obviate the need for frequent dosing to sustain the enzyme activity at a therapeutic
  • controlled release is obtained by appropriate selection of various formulation parameters and Ingredients, Including, e.g., various types of controlled release compositions and coatings.
  • the therapeutic is formulated with appropriate excipients into a pharmaceutical composition that, upon administration, releases the therapeutic in a controlled manner. Examples include single or multiple unit tablet or capsule compositions, oil solutions, suspensions, emulsions, microcapsules, microspheres, molecular complexes, nanoparticJes, patches, and liposomes.
  • compositions of the invention can be incorporated into microspheres, microcapsules, nanoparticles, liposomes, or the like for controlled release.
  • the composition may include suspending, solubilizing, stabilizing, pH-adjusting agents, tonicity adjusting agents, and/or dispersing, agents.
  • the active drug may be incorporated in biocompatible carriers, implants, or infusion devices.
  • Materials for use in the preparation of microspheres and/or microcapsules are, e.g., biodegradable/bioerodible polymers such as polygalactia poly-(isobutyl cyanoacrylate), poly(2-hydroxyethyl-L-glutam ⁇ nine) and, po!y(!actic acid).
  • Biocompatible carriers that may be used when formulating a controlled release parenteral formulation are carbohydrates (e.g., dextrans), proteins (e.g., albumin), lipoproteins, or antibodies.
  • Materials for use in implants can be non-biodegradable (e.g., polydimethyl siloxane) or biodegradable (e.g., poly(caprolactone), poly(lactic acid), poly(glycolic acid) or poly(ortho esters) or combinations thereof).
  • compositions of the invention can comprise various pharmaceutically acceptable salts, ether derivatives, ester derivatives, acid derivatives, and aqueous solubility altering derivatives of the active compound.
  • the present invention can comprise all individual enantiomers, diastereomers, racemates, and other isomer of compounds of the invention.
  • the invention also includes all polymorphs and solvates, such as hydrates and those formed with organic solvents, of this compound. Such isomers, polymorphs, and solvates may be prepared by methods known in the art, such as by regiospeciftc and/or enantioselee ⁇ ve synthesis and resolution, based on the disclosure provided herein.
  • Suitable salts of the compound include, but are not limited to, acid addition salts, such as those made with hydrochloric, hydrobromic, hydroiodic, perchloric, sulfuric, nitric, phosphoric, acetic, propionic, glycolic, lactic pyruvic, malonic, succinic, maleic, fumaric, malic, tartaric, citric, benzoic, carbonic cinnamic, mandelic, methanesulfonic, ethanesulfonic, bydroxyethan ⁇ sulfonie, benezenesulfonic, ⁇ -toluene sulfonic, cyclohexanesulfamic.
  • acid addition salts such as those made with hydrochloric, hydrobromic, hydroiodic, perchloric, sulfuric, nitric, phosphoric, acetic, propionic, glycolic, lactic pyruvic, malonic, succinic, maleic, fumaric,
  • salicyclie, p-aminosalicylic, 2-phenoxybenzoic, and 2-acetoxybenzoic acid salts made with saccharin; alkali metal salts, such as sodium and potassium sails; alkaline earth metal sails, such as calcium and magnesium salts; and salts formed with organic or inorganic ligands, such as quaternary ammonium salts.
  • Additional suitable salts include, but are not limited to, acetate, benzenesulfonate, benzoate, bicarbonate, bisulfate, bitartrale, borate, bromide, calcium edetate, camsylale, carbonate, chloride, clavulanate, citrate, dihydrochloride, edetate, edisylate, estolate, esylate, fumarate, gluceptate, gluconate, glutamate, glycollylarsanilate, hexylresorcinate, hydrabamine, hydrobromide, hydrochloride, hydroxynaphthoate, iodide, isothionate, lactate, lactobionate, laurate, malate, maleate, mandelate, mesylate, methylbromide, methylnitrate, methylsulfate, mucate, napsylate, nitrate, N-methylglucamine ammonium salt
  • a prodrug is a pharmacologically inactive compound that is converted into a pharmacologically active agent by a metabolic transformation. In vivo, a prodrug is acted on by naturally occurring enzyme(s) resulting in liberation of the pharmacologically active agent. Conventional procedures for the selection and preparation of suitable prodrug derivatives are described, for example, in "Design of Prodrugs", ed. H. Bundgaard, Elsevier, 1985.
  • An active metabolite is a compound which results from metabolism of another compound after administration of the latter to a subject. Metabolites can be identified by techniques well-known in the art.
  • kits for the treatment, diagnosis, prophylaxis or prognosis of disease or injury.
  • the kit includes a composition of the invention containing an effective amount of a compound of the invention in unit dosage form.
  • the kit comprises an outer container or package.
  • the kit can comprise a sterile container which contains a therapeutic; such sterile containers can be boxes, ampules, bottles, vials, tubes, bags, pouches, blister-packs, or other suitable container forms known in the art.
  • Such containers can be made of plastic, glass, laminated paper, metal foil, or other materials suitable for holding medicaments.
  • a composition of the invention is provided together with instructions for administering it to a subject. Instructions may include information about the use and effects of the composition. In one embodiment, the instructions will include at least one of the following: description of a composition of the invention, dosage schedule and administration protocols, precautions, warnings, indications, counter-indications, overdosage information, adverse reactions, animal pharmacology, clinical studies, and/or references.
  • the instructions may be printed directly on a container (when present), or as a label applied to the container, or as a separate sheet, pamphlet, card, or folder supplied in, on or with the container. Thus, the instructions may be a separate item in the kit.
  • M 4 N induced TUNEL-positive cell death more efficiently in 67NR than in 4Tl cells.
  • M 4 N suppressed phosphorylation of several PDK-I /PI 3 K/ AKT pathway-related proteins in 67NR but not in 4Tl cells.
  • Transfeetion of PKC ⁇ vectors enhanced cell death induced by M 4 N in both 4Tl and 67NR cells.
  • transfeetion with dominant negative PKC ⁇ (kinase negative) vectors partially suppressed cell death induced by a combination treatment of M 4 N with either Rottlerin or Ly294002.
  • M 4 N was evaluated in a mammary tumors at two different stages metastatic (cell line 4Tl) and nonmetastatic (cell line 67NR). M 4 N is effective to stop the growth of metastatic 4Tl cells but not sufficient to induce cell apoptosis within first 48 hours, although it is able to induce apoptosis in nonmetastatic 67NR cells, (20).
  • M 4 N together with low amounts of additional drugs (drugs that may be correlated with toxicity at higher does) is both efficacious and had essentially nontoxic side effects.
  • additional drugs drugs that may be correlated with toxicity at higher does
  • NDGA derivative is administered daily with a metabolic inhibitor until an effective dosage is achieved in plasma and in the target cells to have the desired therapeutic effect without the toxicity associated with higher dosages that arc necessary when only one drug is administered.
  • agent is meant a polypeptide, peptide, nucleic acid molecule, small molecule, or mimetic.
  • analog is meant an agent having structural or functional homology to a reference agent.
  • cell substrate is meant the cellular or acellular material (e.g., extracellular matrix, polypeptides, peptides, or other molecular components) that is in contact with the cell,
  • control is meant a standard or reference condition.
  • disease is meant any condition or disorder that damages or interferes with the normal function of a cell, tissue, organ or subject,
  • an effective amount is meant the amount of an agent required to ameliorate the symptoms of a disease relative to an untreated patient.
  • the effective amount of an active therapeutic agent used to practice the present invention for the treatment of a disease or injury varies depending upon the manner of administration, the age, body weight, and general health of the subject. Ultimately, the attending clinician will decide the appropriate amount and dosage regimen.
  • fragment is meant a portion of a polypeptide that has at least 50% of the biological activity of the polypeptide from which it is derived. This portion contains, preferably, at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, or 90% of the entire length of the reference nucleic acid molecule or polypeptide.
  • a fragment of a polypeptide or nucleic acid molecule may contain 10, 20, 30, 40, 50, 60, 70, 80, 90, or 100, 200, 300, 400, 500, 600,
  • hypoxia refers to hypoxia inducible factor- 1
  • '"M 4 N refers to Tetra-O-methyl nordihyroguaiaretic acid, EM 1421 or
  • “Lower alkyl” and “lower alkoxy” refer to alkyl and alkoxy groups of 1-6 carbon atoms.
  • modifies is meant alters.
  • an agent that modifies a cell, substrate, or cellular environment produces a biochemical alteration in a component (e.g., polypeptide, nucleotide, or molecular component) of the cell, substrate, or cellular environment.
  • a component e.g., polypeptide, nucleotide, or molecular component
  • MTT refers to 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide.
  • PDK-I indicates 3'-phosphoinositide-dependent protein kinase-1.
  • PBfC refers to phosphatidyl inosito 1-3 -kinase
  • mTOR refers to the mammalian target of rapamycin.
  • the terms "prevent,” “preventing,” “prevention,” “prophylactic treatment” and the like refer to reducing the probability of developing a disorder or condition in a subject, who does not have, but is at risk of or susceptible to developing a disorder or condition.
  • a ''prodrug' is a pharmacologically inactive compound that is converted into a pharmacologically active agent by a metabolic transformation.
  • subject is meant a mammal, including, but not limited to, a human or non- human mammal, such as a bovine, equine, canine, ovine, or feline.
  • therapeutic delivery device any device that provides for the release of a therapeutic agent.
  • exemplary therapeutic delivery devices include osmotic pumps, indwelling catheters, and sustained-release biomaterials.
  • the terms “treat,” treating,” “treatment,” and the like refer to reducing or ameliorating a disorder and/or symptoms associated therewith. It will be appreciated that, although not precluded, treating a disorder or condition does not require that the disorder, condition or symptoms associated therewith be completely eliminated.
  • variant is meant an agent having structural homology to a reference agent but varying from the reference in its biological activity. Variants provided by the invention include optimized amino acid and nucleic acid sequences that are selected using the methods described herein as having one or more desirable characteristics.
  • inhibiting means slowing or stopping the growth of
  • with or “along with” means that the compounds are administered during the same course of treatment, but not necessarily simultaneously. Administration may occur seconds, minutes, or hours apart in time, but will preferably be closely spaced (at least minutes). The compounds should be administered for sufficient duration (e.g. daily) so that an effective dosage is achieved in plasma and in the target tumor cells.
  • FIG. 16 Figure 1. Effect of M 4 N and/or desferoxamine on TUNEL-positive cell death and cell growth in 4Tl and 67NR cells.
  • a and B Either 4Tl (A) or 67NR (B) cells at subconfluent condition were treated with M 4 N (80 ⁇ M) and/or desferoxamine (300 ⁇ M). At 48hrs after treatment the cells were collected and TUNEL assay was performed.
  • C 1x10* 4Tl cells were seeded into 12 well plates. One day later, the cells were treated with different concentrations of MsN. The number of cells was measured by MTT assay at different times points. Data are presented as means (+/ ⁇ ) SD in triplicates.
  • the cells were treated with M 4 N (80 ⁇ M) and/or Desferoxamine (75 or 150 ⁇ M). Five hours later the cells were collected and the western blotting was performed for 4Tl ceils and 67NR cells, 0: without desferoxamine, 75: 75 ⁇ M desferoxamine, 150: 150 ⁇ M desferoxamine, (-): without M 4 N, and M 4 N: M 4 N (80 ⁇ M).
  • PDK-P phospho-PDKl (Ser 24i ), Akt473P: phospho-Akt (Ser 473 ), PRAS40P: phospho-PRAS40 (Thr 246 ), RafP: phospho-Raf (Ser 259 ), and MEK-P: phospho-MEKl/2 (Ser 217 ' 221 ).
  • TUNEL-positiv ⁇ cell death in 4Tl and 67NR cells A: Effect OfM 4 N, Desferoxamine. Rotllerin, Ly294002. and zVAD on TUNEL-positive cell death in 4Tl cells.
  • 4Tl cells at subcontinent condition were treated with M 4 N (80 ⁇ M), Desferoxamine (150 ⁇ M), Rottlerin (5 ⁇ M), or Ly294002 (20 ⁇ M), At 24hrs after treatment the cells were collected and TUNEL assay was performed. When the effect of zVAD (50 ⁇ M) was examined, zVAD had been added 30 minutes before other drugs were added.
  • A&B The expression of the proteins in 4Tl (A) and 67NR (B) detected by the western blotting, The concentration OfM 4 N (M), Rottlerin (Ro) 5 Ly294002 (Ly), and Desferoxamine (D) is 80 ⁇ M, 5 ⁇ M, 20 ⁇ M, and 150 ⁇ M respectively. The combination treatment of Desferoxamine and M 4 N is indicated by DM. Actin was used as control.
  • C&D The cellular localization of total PKC ⁇ (C) and phosphor-PKC ⁇ (Thr 5 ' ' ' 5 ) in 4Tl cells.
  • M 4 N The concentration of M 4 N (M), Rottlerin (Ro), Ly294002 (Ly), and Desferoxamine (Des) is 80 ⁇ M, 5 ⁇ M, 20 ⁇ M, and 150 ⁇ M respectively.
  • the difference between the control and the PKC ⁇ in cells treated with M 4 N and Ly294002 is statistically significant by t- test (2%), B (67NR cells): The difference between the control and the PKC ⁇ wild type in ceils treated with M 4 N is statistically significant by t-test (5%). The difference between the control and the PKC ⁇ K376 mutant in cells treated with M 4 N and Rottlerin is statistically significant by t-test (5%). The difference between the control and the PKC ⁇ K376 mutant in ceils treated with iVljN and L.y294002 is statistically significant by Rest (2%). The difference between the control and the PKC ⁇ in cells treated with M4N and Ly294002 is statistically significant by t-test (1%).
  • FIG. Effect of M 4 N and Rottlerin on the survival time of 4Tl (A) and 67NR (B) tumor-bearing mice.
  • A 5x10 4 4Tl cells were inoculated into fat pads of mammary glands in Balb/c female mice. The injections of either M 4 N (lmg/shot) and Rottlerin (lOO ⁇ g/shot) started 7 days after inoculation of tumors, Drug injections were performed three days a week.
  • B 5xlO 4 67NR cells were inoculated into fat pads of mammary glands in Balb/c female mice.
  • FIG. 8 Effect of M 4 N, Rottlerin, and Ly294002 on TUNEL-positive cell death (A), cell growth (B), and cellular localization of total PKC ⁇ (C) & phosphor-PKC ⁇ (Thr 507 ) (D) in LNCaP cells.
  • concentration of M 4 N (M), Roitl ⁇ rin (R), and Ly294002 (L) is 80 ⁇ M, 5 ⁇ M, and 20 ⁇ M respectively.
  • Cell death was measured at 28hrs after treatment (A), Cell growth assayed by the MTT method was measured at 24hrs after treatment (B).
  • FIG. 10 Metastatic tumors which appeared on the ventral side of nude (nu/nu) mice orthotropicaily implanted with LNCaP tumors with a combination treatment Of M 4 N with Ly294002. There are eight mice with arbitrary designations on the right side of the panels. The pictures were taken after 3 to 6 weeks after inoculation of tumors. The mice were daily injected with M 4 N (Img/shot) and Ly294002 (lOO ⁇ g/shot). Drug injections started 8 days after implantation of tumors. Metastatic tumors are circled by red dots in the pictures.
  • FIG. 11 Effect of combination treatment Of M 4 N and Ly294002 on nude (nu/nu) mice orthotropicaily implanted with LNCaP tumors.
  • LNCaP tumors were orthofropiea ⁇ ly implanted into the vicinity of prostate glawds m male nude mice.
  • Effect of combination treatment Of M 4 N (Img/shot) and Ly294002 (lOO ⁇ g/shot) was examined.
  • Drug injections started 8 days after implantation of tumors. Drugs were administered 7 days a week.
  • A The percentage of mice that have died by the date after tumor inoculation was shown for each group.
  • B Many tumor lesions appeared on the body surface of tumor-bearing mice in one to two weeks after tumor implantation.
  • the size of these tumor lesions was estimated by the calculation described in Materials & Methods. Data are presented as means ( ⁇ +/-) SD. The difference between the control and the combination treatment at both 5 and 6 weeks after tumor implantation is statistically significant by Student's t-test (5%).
  • C The lungs (a & b) and the internal thoracic region facing the lungs (c & d) from the control mice that died of cancer at 7 to 8 weeks after tumor implantation. The lungs (e-g) from the treated mice that were killed at 1 1 weeks after tumor implantation.
  • D The tumors from the treated mice that were killed at 1 1 weeks after tumor transplantation. The tumors were cut by the scalpel so that their inside could be seen, a: the tumor in the prostate region, b-d: the tumors in the abdominal skin.
  • FIG. 12 Effect of combination treatment of M 4 N asid Ly 294002 on metastatis tumors in the hmg and the skin in nude (nu/nu) mice orthotropicaily implanted with LNCaP tumors.
  • A Histological images of the lungs from eight different control mice that died of cancer at 7 to 8 weeks after tumor implantation (a-h). Metastasis lesions (designated as 1 M') exist in six out of eight those mice. Notice that there is not any obvious metastasis lesion in the lung from all four different treated mice thai were killed at 1 1 weeks after tumor implantation (i-1).
  • B Many tumor lesions appeared on the body surface of tumor-bearing mice in one to two weeks after tumor implantation.
  • T8 mouse in this figure is the same one as T8 mouse in figure 3.
  • the mice was daily treated with M 4 N (Img/shot) and Ly294Q02 (lOO ⁇ g/shot). Drug injections started 8 days after implantation of tumors. Rottlerin (lOO ⁇ g/shot) was additionally administered to the mice 10 weeks after the tumor inoculation.
  • FIG. 14 Effect of the combination treatment Of M 4 N with different metabolic modulators on TUNEL-positive cell death in LNCaP cells with different concentrations of drags 24hrs or 48 brs after treatment.
  • LNCaP cells were treated with M 4 N in the combination with either Rottlerin (A) or Ly294002 (B).
  • M 4 N 40 ⁇ M, 60 ⁇ M or 80 ⁇ M
  • Dichloroacetate (10 or 3OmM) or Rapamycin (5, 10, 20, or 30 ⁇ M) were used. Data are presented as means (-*-/-) SD in triplicates (A-F).
  • Figure 15 Effect of combination treatment Of M 4 N with either Ly294002, Rottlerin plus Ly294002 or etoposide on nude (nu/nu) mice orthotropically implanted with LNCaP tumors. LNCaP tumors were orthotropically implanted into the vicinity of prostate giands isi male made mice.
  • A Effect of the combination treatment Of M 4 N with either Ly294002 or Rottlerin plus Ly294002 on the life-span of tumor-bearing mice. The percentage of mice that have died by the date after tumor inoculation was shown for each group.
  • mice in each group were 18, 5, 4, 8, and 9 for the control, Rottlerin alone, M 4 N alone, M 4 N & Ly294002, and M 4 N & Rottlerin & Ly294002 group respectively.
  • Dosages of each injection were Img/shot (for M 4 N) 5 0.2mg/shot (for Ly294002), and O.lmg/shot (for Rottlerin).
  • Drug injections started 3 days after implantation of tumors. Drugs were administered 7 days a week for four weeks. After that drugs were injected once a week.
  • B Effect of the combination treatment Of M 4 N with either Etoposide or Rapamycin on the life-span of tumor-bearing mice.
  • mice inoculation The percentage of mice that have died by the date after tumor inoculation was shown for each group.
  • the numbers of mice in each group were 18, 5, 4, 4, 9 and 5 for the control, Etopside alone, Ly294002 alone, Rapamycin alone, M 4 N alone, M 4 N & Etoposide, and M 4 N & Rapamycin group respectively.
  • Dosages of each injection were Img/shot (for M 4 N), 0.4mg/shot (for Etoposide), and 0.375mg/shot (for Rapamycin).
  • Drug injections started 3 days after implantation of tumors. Drugs were administered 7 days a week for four weeks. After that drugs were injected once a week.
  • C Histological images of the lung from the tumor-bearing mice treated with different methods.
  • M indicates metastasis lesions. Cont, Eto, M 4 N, M4N -S-Ly 5 M 4 N +Ro+Ly, and M 4 N +Eto indicate 'control', 'Etoposide alone', ' M 4 N alone', ' M 4 N and Ly294002 combination', ' M 4 N, Rottlerin, and Ly294002 combination', and ' M 4 N and Etoposide combination'.
  • Fig ⁇ ire 16 Effect of eombsMtl ⁇ E treatment of M 4 N with either Rottierin or Rapamycin in various tumor ceil lines.
  • B TUNEL-positive cell death in various tumor cell lines treated with M 4 N and Rapamycin.
  • the concentration of M 4 N, Rottlerin, and Rapamycin is 80 ⁇ M, either 10 or 20 ⁇ M, and lO ⁇ M respectively.
  • Data are presented as means (+/-) SD in triplicates.
  • Cell Culture Either cell line 4Tl or 67NR was a gift from Dr. Miller (20). The cells were cultured in RPMI 1640 medium supplemented with glucose (14mM), pyruvate (ImM), penicillin (100units/ml) 5 streptomycin (10() ⁇ g/ml), and fetal bovine serum (10%), buffered with 25mM HEPES (pH 7.4). LNCaP human prostate cancer cell line was purchased from American Type Culture Collection (Manassas, VA).
  • the cell line was cultured in RPM ⁇ 1640 medium supplemented with glucose (14mM), pyruvate (ImM), penicillin (100units/ml), streptomycin (lOO ⁇ g/ml), and fetal bovine serum (10%). Drugs were dissolved in dimethyl sulfoxide (DMSOj ai a suitable concentration so that the final concentration of DMSO in the medium was less than 0.1 %, [0134] Reagents: M 4 N was synthesized and supplied by Erimos Pharmaceutical, L. L. C. (Raleigh, NC), according to the method described (87). Rottlerin was from Calbiochem (San Diego, CA). Ly294002 was from Cell Signaling Technology (Danvers, MA).
  • DMSOj ai dimethyl sulfoxide
  • zVAD-fmk was from R&D systems (Minneapolis, MN).
  • Mouse anti-actin antibodies were from Sigma (Saint Louis, MO).
  • Rabbit anti ⁇ HIF, anti-caspas ⁇ 9, anti-phospho-PDK! (Ser 241 ), Akt, phospho-Akt (Ser 473 ) (193H12), Phospho-Raf (Ser 2S9 ), and Phospho-MEKl/2 (Ser 217 ' 7 ' 1 ) antibodies were obtained from Cell Signaling Technology (Danvers, MA).
  • Rabbit anti- phospho-PRAS40 (Thr 246 ) antibody was from Biosource (Camarillo, CA). Desferoxamine mesylate and Etoposide were from Sigma (Saint Louis, MO).
  • mice Female Balb/c mice were obtained from Charles River Laboratories (Wilmington, MA). M 4 N and Rottlerin were dissolved into a solvent, CPE (25/30) solvent system supplied by Erimos Pharmaceutical, L. L. C. (Raleigh, NC) and injected into mice either intravenously or intraperitoneally, T-cell deficient male nude mice (nu/nu) were obtained from Charles River Laboratories (Wilmington, MA). M 4 N and Ly294002 were dissolved into a solvent, CPE (25/30) solvent system supplied by Erimos Pharmaceutical, L.L.C. (Raleigh, NC) and injected into mice intravenously.
  • Tumor inoculation Either 4Tl or 67NR cells were grown as described above. Cells growing subconfluently were collected and resuspended into the tissue culture medium minus fetal bovine serum and antibiotics, After counting the number of the cells, the cell concentration was adjusted. Twenty ⁇ l of the solution containing a selected number of cancer cells (either 5x10 3 or 5xlO 4 cells) was injected into each fat pad of a pair of mammary glands located close to lower limbs. Tumor size was measured by weighing tumors extracted from mouse cadavers.
  • Clonoge ⁇ kity assay for lu ⁇ ig metastasis Clonogenicity assay was done based on the method by Pulanski & Ostrand- Rosenberg (80). At a selected time following tumor inoculation, mice were euthanized, mouse lungs were removed and minced well. Minced lungs were digested in 1 rag/ml collagenase type IV (Sigma, Saint Louis, MO) in phosphate buffered saline without magnesium and calcium (PBS (-)) at 37C for 2hrs. Digested lungs were filtered through 70 ⁇ M cell strainers to remove debris.
  • Pulanski & Ostrand- Rosenberg 80. At a selected time following tumor inoculation, mice were euthanized, mouse lungs were removed and minced well. Minced lungs were digested in 1 rag/ml collagenase type IV (Sigma, Saint Louis, MO) in phosphate buffered saline without magnesium and calcium (PBS (-)) at
  • MTT assay Cells were incubated in PBS (-) supplemented with 5% fetal calf serum and G,5mg/ml MTT, 3-(4,5-Dimethy!thiazol-2-yi)-2,5-diphenyltetrazoliura bromide (Sigma, Saint Louis, MO) for 2hrs. After the incubation, the cells were dissolved in dimethyl sulfoxide and the color of the solutions was measured by a spectrophotometer at wavelengths of 540 and 690nm. The values correlated with viable cell numbers were obtained by subtracting the optical density at 690nm from that at 540nm.
  • Apoptosis assay Simple TLINEL assay was conducted by using TlJNEL apoptosis detection kit (Upstate, Temecula, CA) with some modification. For the evaluation of the effect of PKC ⁇ wild type and dominant negative vectors on TUNEL-positive cell death, we first transfected 4Tl or 67NR cells with these vectors by effectene (Qiagen, Valencia, CA). Wild type PKC ⁇ vector is constructed from wild type PKC ⁇ gene and pEGFP (Promega, Madison, WI).
  • Dominant negative PKC ⁇ vector is constructed from PKC ⁇ gene mutated at a 376th amino acid residue (threonine) and pEGFP (Promega, Madison, WI) This threonine residue is located in ATP -binding domain of protein kinase C ⁇ , and therefore essential for any kinase reaction of the protein so that this mutant protein supposedly function as dominant negative.
  • pEGFP-N3 vector As a control we used pEGFP-N3 vector was used.
  • These vectors contain green fluorescent protein (GFP) gene to be used as a transfection marker.
  • the vectors are kind gifts from Dr. Yuspa (National Institutes of Health, Bethesda, MD) (88).
  • the cells were treated with M 4 N, Rottlerin, or Ly294002. Then 24 hrs after this treatment with these reagents, the cells were fixed with 10% formaldehyde in phosphate buffered saline without calcium and magnesium (PBS (-)). The cells were then incubated with rabbit anti-GFP antibody (MBL International, Woburn, MA) followed by treatment with anti-rabbit IgG antibody conjugated with fluorescein (Promega, Madison, WI).
  • MBL International, Woburn, MA rabbit anti-GFP antibody
  • fluorescein Promega, Madison, WI
  • TUNEL staining was conducted by using TUNEL apoptosis detection kit (Upstate, Temecula, CA) except for using Streptavidin-Alexa Fluor 568 (Molecular Probes, Eugene, OR) instead of Avidine-Rliodamine contained in the kit. Cell death was evaluated by dividing the number of TUNEL-positive cells among the cells positive with anti-GFP staining by the number of these GFP-posiiive cells.
  • the samples were resolved by the standard SDS-polyacrylamide get electrophoresis and transferred to nitrocellulose membrane (Arn ⁇ rsham Bioscienees, Bjorkgatan, Sweden).
  • the membranes were blocked with skim milk, and incubated with primary antibodies at 4°C overnight and then with secondary antibody conjugated with horse radish peroxidase at room temperature for 2hrs.
  • the signals were detected by western blot chemiluminescence reagent plus (New England Nuclear Life Science Products, Boston, MA).
  • LNCaP cells were grown as described above. Cells growing subconfluently were collected and resuspended into the tissue culture medium without fetal bovine serum and antibiotics. After counting the number of the cells, the cell concentration was adjusted using the same medium. After 20 ⁇ l of the medium containing a selected number of cancer cells (about 5x10 7 cells) had been mixed together with the same volume of Matrigel (BD science, Bedford, MA), the combined solution was injected into the skin of nude mice. The tumor tissue growing subcutaneously was used for surgical orthotropic implantation of the tumor into nude mice, according to the method described by Wang et aL (89).
  • the tumor tissue extracted from the skin was excised into pieces of about 2mm diameter, After nude mice were anesthetized by 2,2,2-Tribromoethanol (Aldrich Chemical Co. Inc., Milwaukee, Wf ), a small incision was made at the abdomen of each mouse and a tumor tissue piece was implanted in the neighbor of the prostate of each mouse. Eight days after the operation, the injection of drugs was initiated, and the drugs were injected intravenously every day for the indicated periods
  • M 4 N has undergone Phase I/II clinical trials in patients by intravenous infusion. Two initial reports of these trials showed that M 4 N was able to cause long term stabilization of disease in some patients. However, tumor shrinkage has rarely been seen in patients following M ⁇ N Infusion as only limited amount of the drug can be delivered to tumors in situ by the method used .
  • 4Tl and 67NR cells are the cell lines derived from subpopulations of a mouse breast cancer (20). These two cell lines were characterized by their different behaviors regarding with metastasis.
  • 4Tl or 67NR tumor cells were injected either 4Tl or 67NR tumor cells into fai pads of mammary glands of Balb/c female mice.
  • clonogenicity assay 80
  • Desferoxamine (300 ⁇ M) enhanced cell death induced by M 4 N in either 4Tl or 67NR cells, Since we did not detect many TLJNEL-positive cells in 4Tl cells, we measured the effect of MiN on cell growth in 4Tl cells by MTT assay (Figure 1C). MTT assay showed that M 4 N reduced the cell growth of 4Tl cells depending on the concentration of the drug. Even at 20 ⁇ M of the drug, there was a significant effect of the drag to reduce cell growth, Cell growth stopped at 80 ⁇ M, The effect of M 4 N on cell growth was evident at 42hrs after treatment. [0146] Effect of M 4 N on phosphorylation status of PDK-1/Akt pathway-related proteins in 4Tl and 67NR cells.
  • Desferoxamine induced H ⁇ F stabilization in both 4Tl and 67NR cells as expected and desferoxamine at 150 ⁇ M, a sensor of hypoxic condition, protects HIF very efficiently in both 4Tl and 67NR cells (39).
  • Desferoxamine-mediated HIF induction was slightly suppressed by M 4 N in 4Tl cells while it was instead enhanced by M 4 N in 67NR cells ( Figure 2).
  • Overall the data indicates that M 4 N suppressed PD 1C-].
  • phosphor-PKC ⁇ (Thr 505 ) is distributed rather diffusively in 4Tl cells.
  • M 4 N treatment augmented staining for phosphor-PKC ⁇ (Thr 505 ) in nuclei of the cells ( Figure 4D).
  • Ro also augmented staining for phosphor-PKC ⁇ (Thr 505 ) in nuclei.
  • mice We started to treat mice with drugs seven or eight days after the inoculation of 4Tl or 67NR cells respectively. We injected drugs three days a week after the initiation of drug treatment. Although either M 4 N or Ro alone didn't extend the lifespan of 4Tl -bearing mice, a combination treatment Of M 4 N with Ro extended the life span of these mice (Figure 6A). A similar tendency was found in mice inoculated with 67NR cells as well, As expected, mice overall survived longer with 67NR than 4Tl cell inoculations. While M 4 N or Ro alone didn't have much effect on the life-span of 67NR-bearing mice, a combination treatment of MjN with Ro overall extended the life span of the mice ( Figure
  • LNCaP tumors were orthotropically implanted into the vicinity of the prostate of nude mice, We started to treat the mice with a combination treatment of M 4 N and Ly eight days after the implantation of LNCaP cells. Some tumors started to appear under the skin, especially in the abdomen, for both control and treated mice. While these tumors appeared under the skin all over the body in the control group, they appeared only in the lower abdomen area in the mice treated with the combination ( Figure 9 & 10). All the mice injected with only vehicles died between 38 and 53 days after the tumor implantation. On the contrary, all the mice injected with M 4 N and Ly survived at 76 days after the tumor implantation ( Figure HA).
  • LNCaP tumors were orthotropically implanted into the vicinity of the prostate of nude mice as in the previous studies ( Figures 9 to 13), However, in this experiment we implanted about eight times the volume of tumors as in the previous experiments to detect the difference of efficacy of various combination treatments much faster.
  • mice treated with Etoposide alone died by 24 days after the implantation while all the mice treated with M 4 N alone died by 33 days, The combination treatment of M 4 N with Ly improved the survival of tumor-bearing mice although all the mice eventually died by 51 days (Figure 15A), This is an improvement compared with the survival time in the mice treated with either M 4 N or Ly alone.
  • Phosphorylation of protein kinase C ⁇ (PKC ⁇ ) at Threonine 505 is not a prerequisite for enzymatic activity. J. Biol Chem. 272:6805-6811,
  • Akt/PkB signaling Navigating downstream.

Landscapes

  • Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Epidemiology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)

Abstract

Disclosed is a composition comprising a derivative of NDGA and at least one metabolic modulator. The composition can be in a unit dose form or kit. The composition can comprise at least two metabolic modulators. Also disclosed are methods for achieving cytotoxicity, particularly of rapidly dividing cells such as cancer, by administering a composition of the invention. In various embodiments of the invention subjects with cancer achieve prolonged survival and/or diminution in the size of their malignancies and cancer metastasis.

Description

7gε< F CANCER GROWTH AND METASTASIS USING NORDIHYDROGUAIARETIC ACID DERIVATIVES WITH METABOLIC
MODULATORS
FIELD OF THE INVENTION
[001] This invention relates to the use of nordihydroguaiaretic acid derivatives together with one or more metabolic modulators to treat cancer, prevent metastasis, and prolong the life of a mammal afflicted with a tumor.
BACKGROUND
[002] Carcinogenesis is a multistage event affected by a variety of genetic and epigεnεtic factors and is typified by the outbreak of uncontrolled cell growth originated from different tissues, A universal goal for anticancer research lies in the development of a clinical treatment that is highly effective in curtailment of tumor growth, non-toxic to the host, and is affordable for most patients. Drugs that inhibit targets that are unique to dividing cells, particularly cells dividing in an uncontrolled manner, are an ideal paradigm for chernotherapeutic agents, the greater the specificity to cells that are dividing in an uncontrolled manner the lower the risk of attendant side effects.
[003] Under normal conditions cells in our bodies are involved in a balanced system of programmed growth, division, rest, and death. The regulation of these cellular pathways is essential in order to maintain tissue viability and bodily health. The transition of a healthy cell to a precancerous or cancerous cell is initiated by the disruption of these regulatory pathways. Cancer cells then redirect the cellular systems to allow for uncontrolled cell growth, replication, and/or resistance to programmed cell death {apoptosis). [004] Caspases are one means of inducing apoptosis. Apoptosis is regulated by the inhibitor of apoptosis protein (IAP) family of proteins, through their inhibition of caspase- induced cell death. One of the IAP family members, survivin, is over-expressed in precancerous and cancerous cells, and rarely found in healthy adult cells, By their high survivin expression, tumor cells are prevented from entering the easpase-induced cell death pathway that would lead to their destruction. Survivin is one of the targets currently being tested for anticancer therapy,
[005] Another mechanism by which tumor cells grow uncontrollably is by deregulating their cell cycle process. Cdc2 (cyclin-dependent kinase-1) is one of several kinase proteins controlling cell division and is frequently de-regulated in cancer cells. Cdc2 is also involved in the activation of survivin. In addition, growing tumors require a constant supply of essential nutrients and oxygen. Cancer cells achieve their nutrient needs by secreting Vascular Endothelial Growth Factor (VEGF) to induce new blood vessel formation within the tumor mass.
[006] Proteins such as those mentioned above are involved in the regulation of the signaling pathways that control cell growth, division and death. Some of these proteins are significantly altered in cancer cells. The process of converting the gene sequence on DNA to an RNA message (a process called transcription) that can then be converted to protein (a process called translation) is essential to the regulation of protein production. [007] Cells pass through many checkpoints as they proceed through the eel! cycle. Certain criteria must be met in order to pass each of these checkpoints. In the G2/M transition, the most essential regulator is the cyclin-dependent kinase CDC2. This kinase binds tightly to the regulatory protein cyclin B, and this complex, also called the maturation promoting factor (MPF), is responsible for stimulating a myriad of events that lead to the cell's entry into early prophase (1), Not surprisingly, the loss or deactivation of either component of the MPF will block cellular progression out of G2.
[0Θ8] The expression and activity of the MPF is regulated at different levels. Cyclin B protein levels slowly rise through the Gl and S phases of the cell cycle, peak during the G2 to M phase transition, and drop sharply during mitosis (2). The CDC2 protein, on the other hand, is always present during the cell cycle, although levels rise slightly in the last stages of the G2 phase (3). The activity of the protein is dependent on the association with the appropriate cyclin, as well as on the dephosphorylation of its inhibitory sites by the phosphatase CDC25C (4, 5). It has been shown that the failure of this dephosphorylation initiates G2 arrest in response to DNA damage by radiation or chemical action. Recent evidence also suggests that any remaining active CDC2 may be transported outside the nucleus following DNA damage (6),
[009] Survivin is an inhibitor of apoptosis that is abundantly expressed in many human cancers (7), but not in normal adult human tissue, and is considered a possible modulator of the terminal effector phase of cell death/survival. (8), Survivin is expressed in G2-M in a cell cycle-dependent manner, binding directly to mitotic spindle microtubules. It appears that survivin phosphorylation on Thr34 may be required to maintain ceil viability at cell division (9), and expression of a phosphorylaiion-defecTϊve survivin mutant bas been shown to trigger apoptosis in several human melanoma cell lines ( 10), Phosphorylated survivin acts on the caspase pathway to suppress the formation of caspase-3 and caspase-9.. thereby inhibiting apoptosis. (11) Although compounds that reduce the expression of survivin will be expected to increase the rate of apoptosis and cell death, CDC-2 has been shown to be necessary for survivin phosphorylation (9). In addition, the activation of caspases is a time-dependent event as it occurs slowly, quite often inefficiently.
[010] A number of naturally occurring derivatives of the plant Jignan nordihydroguaiaretic acid (NDGA) have been shown to block viral replication through the inhibition of viral transcription. NDGA is extracted from the resin of the leaves of Larrea tridentata, a desert bush indigenous to the southwestern US and Mexico. Derivatives of NDGA can inhibit the production of HIV (12, 13), HSV (14), and HPV transcripts (15) by the deactivation of their SpI -dependent promoters. Isolation and purification of plant lignans, however, is labor intensive and costly. In anticipation of the possible clinical use of plant lignans in controlling SpI -regulated viral and tumor growth in humans, nine different methylated NDGA activities were synthesized chemically using unmethylated NDGA as the parent substrate in large quantities with low cost (12).
[Oil] Nordihydroguaiaretic acid (M4N5 EM 1421, Terameprocol), is the synthetic telra- methylated derivative of nordihydroguaiaretic acid (tetra-O-methyl nordihydroguaiaretic acid, abbreviated as M4N), The chemical structure of M4N was designed to make it pharmacologically distinct from NDGA. M4N has been shown to possess antiviral (12, 14) and anti-cancer (16) activities in cultured cells, in mouse models (16, 17), and in human xenografts in nude mice (18). M4N causes cell cycle arrest at the G2 phase of the cell cycle probably by suppressing Sp-I regulated cdk expression (16, 19). M4N has been in Phase I clinical trials in patients by intravenous infusion (CLINICAL TRIALS.GOV, A service of U.S. NIH).
[012] Nordihydroguaiaretic acid (NDGA) derivatives such as M4N suppress SpI regulated transcription of viral genes, by deactivation of SpI -dependent promoters. SPl also affects expression of many growth-related genes. Cdc2 (also referred to as CDKl) and cyclin B interact to allow cells to move from the G2 phase of celi division to mitosis. M4N blocks the transcription of Cdc2, and thus blocks cell division. The Sp 1 protein on promoter of CDC2 chromatin is replaced following M4N treatment in vivo.
[013] M4N is able to induce cell cycle arrest in mammalian cell lines; M4N is a transcription inhibitor. It selectively reduces transcription of growth related genes that have promoters controlled by the SpI factor, such as cdc2, survivin and VEGF. By blocking production of cdc2, and VEGF, M4N inhibits tumor growth and starves nmiors by restricting growth of their blood supply. [014] MJN has been shown to arrest growth of a variety of human cells in vitro, the majority of which are part of the NCI panel of 60 cancer cell lines, including solid tumor cell lines (bladder, breast, colorectal, liver, lung, ovarian, pancreatic, prostate and cervical carcinomas), and erythroleukemia cells. In vivo, M4N also decreases tumor cell growth and exhibits antitumor activity in a large number of tumor xenograft models, including human bladder, breast, colorectal, liver, ovarian, pancreatic, prostate and cervical carcinomas, and erythroleukemia, without apparent toxicity. MjN has a broad spectrum of activity in anticancer therapy, having affects on Cdc2, HIF- lα, MDRl, VEGF and survivin. For example, M4N induces apopiosis and reduces cdc2 protein levels in human oral cancers. M4N also appears to reduce survivin levels in these cancers. Administered systemically, M4N was also shown to inhibit xenografted human tumors MCF-7, Hep3b, LNCaP, HT-29, and Iv562, Although none of the xenograft tumors were fully eradicated,
[015] M4N does not appear toxic to animals. For example, M4N retention in mouse organs following oral administration has been studied after short term and long term feeding, the results showed essentially no toxic effects even at concentrations high as 906 μg/g of tissue. On daily (1 rng/day) IV injection of M4N for days, M4N accumulated in blood and tumors to levels above 1 mM in nude mice carrying human tumor xenografts. [016] By use of gene array studies with 9600 expressed genes. Applicants previously found products of most SpI regulated genes remained at similar levels, and not affected by the drug treatment of cervical cancer cells C3 in culture..
[017] M4N has some favorable therapeutic qualities, in that it exhibits efficacy against several tumors, by inhibiting cell growth. However, in human clinical trials, treatment with M4N does not generally eradicate disease, and upon cessation of treatment with M4N tumors are capable of growing back. Accordingly, there is a need to identify ways to boost the efficacy of the M4N when the type of cancer is aggressive, metastatic or when M4N as a single drug in low concentration is not enough to induce rapid apoptosis of such type of cancers.
[018] This application claims priority to U.S. Provisional Patent Application 60/010,371 , filed January 8, 2008, and U.S. Provisional Patent Application 61/191,827, filed September 12, 2008, each of which is incorporated by reference in its entirety. [019] As set forth herein, M4N and related derivatives of nordihydroguaiaretic acid have been used in combination with metabolic inhibitors to produce excellent results in tumor treatment, including prolonged patient survival as well as cytotoxicity to malignant cells. [020] Accordingly, it is one object to provide a pharmaceutical composition comprising an effective amount of nordihydroguaiaretic acid (NfDGA) or a derivative thereof of formula
Figure imgf000007_0001
wherein Ri, R2, R3 and R4 independently represent hydroxy, a straight or branched chain lower alkyl or alkoxy, an amino acid residue, a substituted amino acid residue, a nitrogen- containing 5- or 6-membered heterocyclic ring or a saccharide residue; the amino acid residue, substituted amino acid residue, nitrogen-containing 5 or 6 membered heterocyclic ring or saccharide residue being optionally joined to the phenyl ring by a linker of an oxygen atom and 1-10 carbon atoms, and an effective amount of a metabolic modulator.
[021] In one specific embodiment, the pharmaceutical composition comprises the NDGA derivative tetra-ø~methyl nordihydroguaiaretic acid (M4N). In another specific embodiment, the pharmaceutical composition comprises the NDGA derivative maltose M3N (90). In other specific embodiments, Rj, R2, R3 and R4 are identical and represent straight or branched chain lower alkoxy groups or naturally occurring amino acid residues.
[022] The metabolic modulator can be selected, for example from the group consisting of an inhibitor of the PDK-1/PI3K/AKT pathway, an inhibitor of the PKCδ/topoisomerase ϊϊα pathway, an inhibitor of mitochondrial permeability transition, and an inhibitor of the mitogen-activated protein kinase (MAPK/RAS) pathway, or any other metabolic modulators which can systematically induce translocation of phosphoryϊated PC&δ to the nucleus of tumor cells. Examples of such metabolic inhibitors are Ly294002, etoposide, Rottlerin, dichloroacetate, cβtuximab, trastuzurnab, bevacizumab and rapamyein. [023] In certain preferred embodiments, the metabolic inhibitor is etoposide, rapamycin, rottlerin and/or water soluble derivatives thereof, such as temsirolimus, everolimus and other selected molecular targeting agents in clinical development (Nature Reviews Drug Discovery vol 5, p 650, 2006).
[024] Also provided is a method of treating a tumor, comprising administering to a mammal in need of treatment an effective amount of the above-described pharmaceutical composition. The mammal can be any mammal afflicted with a tumor amenable to treatment, for example a human, nonhuman primate, cat, dog, mouse, etc. The tumor may be a solid or hematological tumor, benign or malignant (metastatic or nonmεtastatic), such as, for example, breast, liver, prostate, cervical, ovarian, colon, brain, pancreatic, bladder esophagus, gut, head and neck, kidney, melanoma, stomach, testes, thyroid, uterine and lung cancers, leukemias and lymphomas, such as acute myelogenous leukemia, acute or chronic lymphocytic leukemia, Hodgkin's and non-Hodgkin lymphoma, and myelomas. Persons of skill in the art will be able to determine by routine experimentation the types of tumors that are amenable to treatment. The treatment method is particularly suitable for treatment of metastatic and nonmetastatic cancer.
[025] Also provided is a method of preventing or inhibiting tumor growth in an animal, said method comprising administering an effective amount of nordihydroguaiaretic acid (NDGA) or a derivative thereof of formula I:
Figure imgf000008_0001
wherein Rj5 R2, R3 and R4 independently represent hydroxy, a straight or branched chain lower alkyl or alkoxy, an amino acid residue, a substituted amino acid residue, a nitrogen- containing 5- or 6~membered heterocyclic ring or a saccharide residue; the amino acid residue, substituted amino acid residue, nitrogen-containing 5 or 6 membered heterocyclic ring or saccharide residue being optionally joined to the phenyl ring by a linker of an oxygen atom and 1-10 carbon atoms, and an effective amount of a metabolic modulator. [026] In one specific embodiment, the NDGA derivative is tetra-o-methyl nordihydroguaiaretic acid (M4N). In another specific embodiment, the N DGA derivative is maltose M3N (90). In other specific embodiments, R1, R2, R3 and R4 are identical and represent straight or branched chain lower alkoxy groups or naturally occurring amino acid residues.
[027] The mammal can be any mammal afflicted with a tumor amenable to treatment, for example a human, nonhuman primate, cat, dog, mouse, etc. The metabolic inhibitor is selected from the group consisting of an inhibitor of the PDK-1/PI3K/AKT pathway, an inhibitor of the PKCδ/topoisomerase Ilα pathway, an inhibitor of mitochondrial permeability transition, and an inhibitor of the mitogen-activated protein kinase (MAPK/RAS) pathway. For example, the metabolic inhibitor is selected from the group consisting of Ly294002, etoposide, Rottlerin, dichloroacetate, cetuximab, trastuzumab, bevacizumab and rapamycin. Etoposide, rapamycin, and water soluble derivatives thereof are expected to be particularly effective.
[028] The tumor may be a solid or hematological tumor, benign or malignant (metastatic or nonmetastatic), such as, for example, breast, prostate, cervical, ovarian, colon, brain, pancreatic and lung cancers, leukemias and lymphomas, and others mentioned hereinabove. Persons of skill in the art will be able to determine by routine experimentation the types of tumors that are amenable to treatment. The treatment method is particularly suitable for treatment of metastatic and nonmetastatic cancer.
[029] Also provided is a method of preventing tumor metastasis in a mammal, said method comprising administering an effective amount of nordihydroguaiaretic acid (NDGA) or a derivative thereof of formula ϊ:
Figure imgf000009_0001
wherein R;, R2, R.3 and R4 independently represent hydroxy, a straight or branched chain lower aikyl or alkoxy s an amino acid residue, a substituted amino acid residue and a saccharide residue; the amino acid residue, substituted amino acid residue or saccharide residue being optionally joined to the phenyl ring by a Maker of an oxygen atom and 1-10 carbon atoms; and an effective amount of a metabolic modulator.
[030] In one specific embodiment, the NDGA derivative is ietra~o~methyl nordihydroguaiaretic acid (M4N), In another specific embodiment, the NDCJA derivative is maltose M3N (90). In other specific embodiments, Rj, R:,, R3 and R4 are identical and represent straight or branched chain lower alkoxy groups or naturally occurring amino acid residues.
[031] The mammal can be any mammal afflicted with a tumor amenable to treatment, for example a human, nonhuman primate, cat, dog, mouse, etc, The metabolic inhibitor is selected from the group consisting of an inhibitor of the PDK-1/PI3K/AKT pathway, an inhibitor of the PKCδ/topoisomerase Ilα pathway, an inhibitor of mitochondrial permeability transition, and an inhibitor of the mitogen-activated protein kinase (MAPK/RAS) pathway.
For example, the metabolic inhibitor is selected from the group consisting of Ly294002, efoposide, Rottlεrin, dichloroacetate, cetuxtmab, trastuzuπiab, bevaeizumab and rapamycin.
Etoposide, rapamycin, and water soluble derivatives thereof are expected to be particularly effective.
[032] The tumor may be a solid or hematological tumor, such as, for example, breast, prostate, cervical, ovarian, colon, brain, pancreatic and lung cancers, leukemias and lymphomas, and others mentioned hereinabove, Persons of skill in the art will be able to determine by routine experimentation the types of tumors that are amenable to treatment.
[033] Treatment may be administered alone, or as an adjuvant to surgery, e.g. before surgery, for example, to reduce tumor size, and/or following surgery to reduce the possibility of metastases, e.g. by inhibition of the growth and migration of circulating tumor cells through the blood stream,
[034] The invention also provides a method of prolonging the life of a mammal having a malignant tumor, said method comprising administering to the mammal an effective amount of nordihydroguaiaretic acid (NDGA) or a derivative thereof of formula I:
Figure imgf000010_0001
(I) wherein R-., R,J, R3 and R4 independently represent hydroxy, a straight or branched chain lower alkyS or alkoxy, an amino acid residue, a substituted amino acid residue, and a saccharide residue; the amino acid residue, substituted amino acid residue or saccharide residue being optionally joined to the phenyl ring by a linker of an oxygen atom and 1-10 carbon atoms; and an effective amount of a metabolic modulator.
[035] In one specific embodiment, the NDGA derivative is tetra-o-methyl nofdihydroguaiaretic acid (M4N), In another specific embodiment, the NDGA derivative is maltose M3N (90). In other specific embodiments, Ri, R2, R3 and R4 are identical and represent straight or branched chain lower alkoxy groups or naturally occurring amino acid residues.
[036] The mammal can be any mammal afflicted with a tumor amenable to treatment, for example a human, nonhuman primate, cat, dog, mouse, etc. The metabolic inhibitor is selected from the group consisting of an inhibitor of the PDK-I /PB K/ AKT pathway, an inhibitor of the PKCδ/topoisomerase ϊϊα pathway, an inhibitor of mitochondrial permeability transition, and an inhibitor of the mitogen-activated protein kinase (MAPIC7RAS) pathway.
For example, the metabolic inhibitor is selected from the group consisting of Ly294002, etoposide, Rottlerin, dichloroacetate, cetuximab, trastuzumab, bevacizumab and rapamycin.
Etoposide, rapamycin, and water soluble derivatives thereof are expected to be particularly effective.
[037] The tumor may be a solid or hematological tumor, such as, for example, breast, prostate, cervical, ovarian, colon, brain, pancreatic and lung cancers, leukemias and lymphomas, and other tumors mentioned hereinabove. Persons of skill in the art will be able to determine by routine experimentation the types of tumors that are amenable to treatment.
Figure imgf000011_0001
[039] Pharmaceutical compositions in accordance with the invention, are useful for diagnosis, prognosis , prophylaxis or treatment of a condition. Accordingly, compositions in accordance with the invention are useful as a drug or as information for structural modification of existing compounds, e.g., by rational drug design. Compounds and methods of the invention are useful for screening compounds having an effect on a variety of conditions. [040] For therapeutic uses, the compositions or agents identified using the methods disclosed herein may be administered systemically, for example, formulated in a pharmaceutically-acceptable buffer such as physiological saline. Preferable routes of administration include, for example, subcutaneous, intravenous, intraperitoneally., intramuscular, or intradermal injections that provide continuous, sustained levels of the drug in the patient, Treatment of human patients or other animals are generally carried out using a therapeutically effective amount of a therapeutic of the invention in a physiologically- acceptable carrier, Suitable carriers and their formulation are described, for example, in Remington's Pharmaceutical Sciences by E. W. Martin.
[041] The amount of the therapeutic agent to be administered varies depending upon the manner of administration, the age and body weight of the subject/patient, and with the subject's symptoms and condition. A compound is administered at a dosage that best achieves medical goals with the fewest corresponding side effects. [042] Administration
[043] The pharmaceutical compositions of this invention including biologically active fragments, variants, or analogs thereof, can be administered by any suitable routes including intracranial, intracerebral, intraventricular, intrathecal, intraspinal, oral, topical, rectal, transdermal, subcutaneous, intravenous, intramuscular, intranasal, and the like. In one embodiment, the compositions are added to a retained physiological fluid, such as cerebrospinal fluid, blood, or synovial fluid. The compositions of the invention can be amenable to direct injection or infusion at a site of disease or injury. [044] Compositions for parenteral use may be provided in unit dosage forms (e.g., in single-dose ampules), or in vials containing several doses and in which a suitable preservative may be added. The composition may be in the form of a solution, a suspension, an emulsion, an infusion device, or a deliver}' device for implantation, or it may be presented as a dry powder to be reconstituted with water or another suitable vehicle before use. The composition may include suitable parenterally acceptable carriers and/or excipients. [045] In one approach, a therapeutic of the invention is provided within an implant, such as an osmotic pump, or in a graft comprising appropriately transformed cells. Methods of introduction may also be provided by rechargeable or biodegradable devices. Various slow release polymeric devices have been developed and tested for the controlled delivery of drugs, including proteinacious biopharmaceuticals. A variety of biocompatible polymers (including hydrogεls), including both biodegradable and non-degradable polymers, can be used to form an implant for the sustained release of a bioactive factor at a particular target site.
[046] Generally, the amount of administered agent of the invention (dosage) will be empirically determined in accordance with information and protocols known in the art.
Typically agents are administered in the range of about 10 μg/kg to 100 mg/kg of the recipient. Other additives may be included, such as stabilizers, bactericides, and anti-fungals.
These additives will be present in conventional amounts,
[047] Exemplary dosages to be considered are shown in Table 1.
Table 1
Estimation of the amounts of Terameproeol, Etoposide, and Rapamycin to be considered in patient treatment
A. Terameproeol
Km factor*
Mouse 1 mg/day 30.3 mg/kg 90.3 mg/m2
Human 171.5 mg/day 2.45 mg/kg 37 90.3 mg/m2 As mouse
859.6 mg/day 12.28 mg/kg 37 454.5 mg/m2 5x mouse
B. Etoposide
Mouse 0.4 mg/day 12.1 mg/kg 36.3 mg/m
Human 68.6 mg/day 0.98 mg/kg 37 36.3 mg/m2 As mouse
343 mg/day 4.9 mg/kg 37 181.5 mg/m2 5 x mouse
C, Rapamycin
Mouse 0.375 mg/day 11.1 mg/kg 3 33.3 mg/nr
Human 63 mg/day 0.9 mg/kg 37 33.3 mg/m2 As mouse
315 mg/day 4.5 mg/kg 37 166.5 mg/m2 5x mouse
Mouse weight: 33 g Human weight: 70 kg
*Reference: Dose Translation from Animal to Human Studies
S. R. Shaw, M. Nihal, and N. Ahmad
The FASEB Journal, Vol. 22 p.659-661, 2007
[048] Columns 2 and 3 of Table 1 show effective dosages administered in mice in experiments as described herein, and administered to humans in clinical trials. Column 4 shows Km factor for conversion to patient treatment dosage. Column 5 gives the calculated amount for administration to patients. In general it is expected that blood levels of ,5 to 10 mM of M4N, preferably 1-5 mM, should be achieved for effective patient treatment. Persons of skill in the art will appreciate that the dosage is expected to vary depending on the protocol used for drug administration.
[049] The administration of a compound of the invention may be by any suitable means that results in a concentration of the therapeutic that, combined with other components, is effective in ameliorating, reducing, or stabilizing a deficit or disorder. The compound may be contained in any appropriate amount in any suitable carrier substance, and is generally present in an amount of 1-95% by weight of the total weight of the composition. The composition may be provided in a dosage form that is suitable for parenteral (e.g., subcutaneously, intravenously, intramuscularly, or intraperitoneally) administration route. The pharmaceutical compositions may be formulated according to conventional pharmaceutical practice (see, e.g., Remington: The Science and Practice of Pharmacy (20th ed.), ed. A. R. Gennaro, Lippincott Williams & Wilkins, 2000 and Encyclopedia of Pharmaceutical Technology, eds. J. Swarbrick and J. C. Boylan, 1988-1999, Marcel Dekker, New York).
[050] Formulation of Pharmaceutical Compositions
[051] As noted above, compositions of the invention can be administered parenterally by injection, infusion or implantation (subcutaneous, intravenous, intramuscular, intraperitoneal, or the like) in dosage forms, formulations, or via suitable delivery devices or implants containing conventional, non-toxic pharmaceutically acceptable carriers and adjuvants. The formulation and preparation of such compositions are well known to those skilled in the art of pharmaceutical formulation. Formulations can be found in Remington: The Science and Practice of Pharmacy, cited herein.
[052] For example, pharmaceutical compositions according to the invention may be in the form suitable for sterile injection. To prepare such a composition, the compositions(s) are dissolved or suspended in a parenterally acceptable liquid vehicle. Among acceptable vehicles and solvents that may be employed are water, water adjusted to a suitable pH by addition of an appropriate amount of hydrochloric acid, sodium hydroxide or a suitable buffer, 1,3-butanediol, Ringer's solution, and isotonic sodium chloride solution and dextrose solution. The aqueous formulation may also contain one or more preservatives (e.g., methyl, ethyl or n-propyl p-hydroxybenzoate).
1053] Suitable dosage forms can be formulated for, but are not limited to, oral, rectal, sub-lingual, mucosal, nasal, ophthalmic, subcutaneous, intramuscular, intravenous, transdermal, spinal, intrathecal, intra-articular, intra-artεrial. sub-arachinoid, bronchial, lymphatic, and intra-uterille administration, and other dosage forms for systemic delivery of active ingredients. In a preferred embodiment, the dosage form is suitable for injection or intravenous administration.
[054] To prepare such pharmaceutical dosage forms, one or more of the aforementioned compounds are intimately admixed with a pharmaceutical carrier according to conventional pharmaceutical compounding techniques. The carrier may take a wide variety of forms depending on the form of preparation desired for administration.
[055] For parenteral formulations, the carrier will usually comprise sterile water, though other ingredients, for example, ingredients that aid solubility or for preservation, may be included. Injectable solutions may also be prepared in which case appropriate stabilizing agents may be employed.
[056] In preparing the compositions in oral dosage form, any of the usual pharmaceutical media may be employed. Thus, for liquid oral preparations, such as, for example, suspensions, elixirs and solutions, suitable carriers and additives include water, glycols, oils, alcohols, flavoring agents, preservatives, coloring agents and the like. For solid oral preparations such as, for example, powders, capsules and tablets, suitable carriers and additives include starches, sugars, diluents, granulating agents, lubricants, binders, disintegrating agents and the like. Due to their ease in administration, tablets and capsules represent the most advantageous oral dosage unit form, [f desired, tablets may be sugar coated or enteric coated by standard techniques.
[057] In some applications, it may be advantageous to utilize the active agent in a
"vectorized" form, such as by encapsulation of the active agent in a liposome or other encapsulant medium, or by fixation of the active agent, e.g., by covalent bonding, chelation, or associative coordination, on a suitable biomoleeule, such as those selected from proteins, lipoproteins, glycoproteins, and polysaccharides.
[058] Methods in accordance with the present invention using formulations suitable for oral administration may be presented as discrete units such as capsules, cachets, tablets, or lozenges, each containing a predetermined amount of the active ingredient as a powder or granules. Optionally, a suspension in an aqueous liquor or a non-aquεous liquid may be employed, such as a syrup, an elixir, an emulsion, or a draught.
|l)59] Formulations for oral use include tablets containing active ingredient(s) of the invention in a mixture with pharmaceutically acceptable excipients, Such formulations are known to the skilled artisan. Excipients may be, for example, inert diluents or fillers (e.g., sucrose, sorbitol, sugar, manniiol, mieroeryslallϊne cellulose, starches including potato starch. calcium carbonate, sodium chloride, lactose, calcium phosphate, calcium sulfate, or sodium phosphate); granulating and disintegrating agents (e.g., cellulose derivatives including microcrystalline cellulose, starches including potato starch, croscarmellose sodium, alginates, or alginic acid); binding agents (e.g., sucrose, glucose, sorbitol, acacia, alginic acid, sodium alginate, gelatin, starch, prege latinized starch, microcrystalline cellulose, magnesium aluminum silicate, carboxymethylcellulose sodium, methylcellulose, hydroxypropyl methylcellulose, ethylcellulose, polyvinylpyrrolidone, or polyethylene glycol); and lubricating agents, glidants, and antiadhesives (e.g., magnesium stearatε, zinc stεarate, stearic acid, silicas, hydrogenated vegetable oils, or talc). Other pharmaceutically acceptable excipients can be colorants, flavoring agents, plasticizers, humectants, buffering agents, and the like.
[060] As appropriate, a tablet may be made by compression or molding, or wet granulation, optionally with one or more accessory ingredients. Compressed tablets may be prepared by compressing in a suitable machine, with the active compound being in a free- flowing form such as a powder or granules which optionally is mixed with a binder, disintegrant, lubricant, inert diluent, surface active agent, or discharging agent. Molded tablets comprised of a mixture of the powdered active compound with a suitable carrier may be made by molding in a suitable machine.
[061] As appropriate, a syrup may be made by adding the active compound to a concentrated aqueous solution of a sugar, for example sucrose, to which may also be added any accessory ingredient(s). Such accessory ingredient(s) may include flavorings, suitable preservative, agents to retard crystallization of the sugar, and agents to increase the solubility of any other ingredient, such as a polyhydroxy alcohol, for example glycerol or sorbitol. [062] Formulations suitable for parenteral administration usually comprise a sterile aqueous preparation of the active compound, which preferably is isotonic with the blood of the recipient (e.g., physiological saline solution). Such formulations may include suspending agents and thickening agents and liposomes or other microparticulate systems which are designed to target the compound to blood components or one or more organs. The formulations may be presented in unit-dose or multi-dose form.
[1)63] Parenteral administration may comprise any suitable form of systemic delivery or localized delivery. Administration may for example be intravenous, intra-arterial, intrathecal, intramuscular, subcutaneous, intramuscular, intra-abdominal (e.g., intraperitoneal), etc., and may be effected by infusion pumps (external or implantable) or any other suitable means appropriate to the desired administration modality. [064] Nasal and other mucosal spray formulations (e.g. inhalable forms) can comprise purified aqueous solutions of the active compounds with preservative agents and isotonic agents. Such formulations are preferably adjusted to a pH and isotonic state compatible with the nasal or other mucous membranes. Alternatively, they can be in the form of finely divided solid powders suspended in a gas carrier. Such formulations may be delivered by any suitable means or method, e.g., by nebulizer, atomizer, metered dose inhaler, or the like. [065] Formulations for rectal administration may be presented as a suppository with a suitable carrier such as cocoa butter, hydrogenated fats, or hydrogenated fatty carboxylic acids.
[066] Transdermal formulations may be prepared by incorporating the active agent in a thixotropic or gelatinous carrier such as a cellulosic medium, e.g., methyl cellulose or hydroxyethyl cellulose, with the resulting formulation then being packed in a transdermal device adapted to be secured in dermal contact with the skin of a wearer. [067] In addition to the aforementioned ingredients, formulations of the invention may further include one or more accessory ingredient(s) selected from diluents, buffers, flavoring agents, binders, disintegrants, surface active agents, thickeners, lubricants, preservatives (including antioxidants), and the like.
[068] A formulation of the present invention can have immediate release, sustained release, delayεd-onset release or any other release profile known to one skilled in the art. [069] Pharmaceutical compositions according to the invention may be formulated to release the active compound substantially immediately upon administration or at any predetermined time or time period after administration. The latter types of compositions are generally known as controlled release formulations, which include (i) formulations that create a substantially constant concentration of the drug within the bodv over an extended period of time; (ii) formulations that after a predetermined lag time create a substantially constant concentration of the drug within the body over an extended period of time; (iii) formulations that sustain action during a predetermined time period by maintaining a relatively, constant, effective level in the body with concomitant minimization of undesirable side effects associated with fluctuations in the plasma level of the active substance (sawtooth kinetic pattern); (iv) formulations that localize action by, e.g., spatial placement of a controlled release composition adjacent to or in the central nervous system or cerebrospinal fluid; (v) formulations that allow for convenient dosing, such that doses are administered, for example, once every one or two weeks; and (vi) formulations that target the site of a pathology. For some applications, controlled release formulations obviate the need for frequent dosing to sustain the enzyme activity at a therapeutic level.
[070] Any of a number of strategies can be pursued in order to obtain controlled release in which the rate of release outweighs the rate of metabolism of the compound in question. In one example, controlled release is obtained by appropriate selection of various formulation parameters and Ingredients, Including, e.g., various types of controlled release compositions and coatings. Thus, the therapeutic is formulated with appropriate excipients into a pharmaceutical composition that, upon administration, releases the therapeutic in a controlled manner. Examples include single or multiple unit tablet or capsule compositions, oil solutions, suspensions, emulsions, microcapsules, microspheres, molecular complexes, nanoparticJes, patches, and liposomes.
[071] The compositions of the invention can be incorporated into microspheres, microcapsules, nanoparticles, liposomes, or the like for controlled release. Furthermore, the composition may include suspending, solubilizing, stabilizing, pH-adjusting agents, tonicity adjusting agents, and/or dispersing, agents. Alternatively, the active drug may be incorporated in biocompatible carriers, implants, or infusion devices. [072] Materials for use in the preparation of microspheres and/or microcapsules are, e.g., biodegradable/bioerodible polymers such as polygalactia poly-(isobutyl cyanoacrylate), poly(2-hydroxyethyl-L-glutam~nine) and, po!y(!actic acid). Biocompatible carriers that may be used when formulating a controlled release parenteral formulation are carbohydrates (e.g., dextrans), proteins (e.g., albumin), lipoproteins, or antibodies. Materials for use in implants can be non-biodegradable (e.g., polydimethyl siloxane) or biodegradable (e.g., poly(caprolactone), poly(lactic acid), poly(glycolic acid) or poly(ortho esters) or combinations thereof). [073] SjiHsjΛnd..Per|yaftves;
[074] Compositions of the invention can comprise various pharmaceutically acceptable salts, ether derivatives, ester derivatives, acid derivatives, and aqueous solubility altering derivatives of the active compound. The present invention can comprise all individual enantiomers, diastereomers, racemates, and other isomer of compounds of the invention. The invention also includes all polymorphs and solvates, such as hydrates and those formed with organic solvents, of this compound. Such isomers, polymorphs, and solvates may be prepared by methods known in the art, such as by regiospeciftc and/or enantioseleeϋve synthesis and resolution, based on the disclosure provided herein. [075] Suitable salts of the compound include, but are not limited to, acid addition salts, such as those made with hydrochloric, hydrobromic, hydroiodic, perchloric, sulfuric, nitric, phosphoric, acetic, propionic, glycolic, lactic pyruvic, malonic, succinic, maleic, fumaric, malic, tartaric, citric, benzoic, carbonic cinnamic, mandelic, methanesulfonic, ethanesulfonic, bydroxyethanεsulfonie, benezenesulfonic, ρ-toluene sulfonic, cyclohexanesulfamic. salicyclie, p-aminosalicylic, 2-phenoxybenzoic, and 2-acetoxybenzoic acid; salts made with saccharin; alkali metal salts, such as sodium and potassium sails; alkaline earth metal sails, such as calcium and magnesium salts; and salts formed with organic or inorganic ligands, such as quaternary ammonium salts.
[076] Additional suitable salts include, but are not limited to, acetate, benzenesulfonate, benzoate, bicarbonate, bisulfate, bitartrale, borate, bromide, calcium edetate, camsylale, carbonate, chloride, clavulanate, citrate, dihydrochloride, edetate, edisylate, estolate, esylate, fumarate, gluceptate, gluconate, glutamate, glycollylarsanilate, hexylresorcinate, hydrabamine, hydrobromide, hydrochloride, hydroxynaphthoate, iodide, isothionate, lactate, lactobionate, laurate, malate, maleate, mandelate, mesylate, methylbromide, methylnitrate, methylsulfate, mucate, napsylate, nitrate, N-methylglucamine ammonium salt, oleate, pamoate (εmbonate), palmitate, pantothenate, phosphate/diphosphatε, poiygalacturonatε, salicylate, stearate, sulfate, subacelate, succinate, tannale, tartrate, teoclate, tosylate, triεthiodide and valerate salts of the compound of the present invention. [077] Prodrugs and active metabolites of compounds of the invention are also within the scope of the invention.
[078] A prodrug is a pharmacologically inactive compound that is converted into a pharmacologically active agent by a metabolic transformation. In vivo, a prodrug is acted on by naturally occurring enzyme(s) resulting in liberation of the pharmacologically active agent. Conventional procedures for the selection and preparation of suitable prodrug derivatives are described, for example, in "Design of Prodrugs", ed. H. Bundgaard, Elsevier, 1985.
[079] An active metabolite is a compound which results from metabolism of another compound after administration of the latter to a subject. Metabolites can be identified by techniques well-known in the art.
[080] The invention also comprises kits, e.g., for the treatment, diagnosis, prophylaxis or prognosis of disease or injury. In one embodiment, the kit includes a composition of the invention containing an effective amount of a compound of the invention in unit dosage form. In some embodiments, the kit comprises an outer container or package. The kit can comprise a sterile container which contains a therapeutic; such sterile containers can be boxes, ampules, bottles, vials, tubes, bags, pouches, blister-packs, or other suitable container forms known in the art. Such containers can be made of plastic, glass, laminated paper, metal foil, or other materials suitable for holding medicaments.
[081] In certain kit embodiments, a composition of the invention is provided together with instructions for administering it to a subject. Instructions may include information about the use and effects of the composition. In one embodiment, the instructions will include at least one of the following: description of a composition of the invention, dosage schedule and administration protocols, precautions, warnings, indications, counter-indications, overdosage information, adverse reactions, animal pharmacology, clinical studies, and/or references. [082] The instructions may be printed directly on a container (when present), or as a label applied to the container, or as a separate sheet, pamphlet, card, or folder supplied in, on or with the container. Thus, the instructions may be a separate item in the kit. or be imprinted embossed, molded or otherwise affixed to another item in the kit; instructions may be. printed on an outer container and also included as an insert item in the kit. [083] The effect Of M4N was studied for 4Tl and 67NR cell lines. These cell lines are derived from subpopulations of a single mouse breast cancer although 4Tl is far more metastatic than 67NR cells.
[084] M4N induced TUNEL-positive cell death more efficiently in 67NR than in 4Tl cells. M4N suppressed phosphorylation of several PDK-I /PI 3 K/ AKT pathway-related proteins in 67NR but not in 4Tl cells.
[085] We found that manipulations of PDK-1/PI3K/AKT pathway by other drugs could modulate the effect of M4N on 4Tl cells. Both Ly294002 (phosphatidyl inositol 3~kinasc inhibitor) and roϊtlcrin (an inhibitor with broad specificity), were found to promote ceil death synergisticaily with M4N in both 411 and 67NfR cells in tissue cultures, Treatment of 4Tl or 67NR tumor-bearing Balb/c female mice with MΛN did not extend the life-span of these mice. However, combination treatment Of M4N with rottlerin extends the life-span of these mice. Additionally combination treatment of M4N with rottlerin reduced lung metastasis of 4Tl cells, measured by the clonogenicity assay. The data indicates possible clinical applications of combination treatment of MiN with rottlerin and Ly294ϋO2 for cancer therapy,
[QBb] The effect of five metabolic modulators (Rottlerin, Ly294Q02, Etoposide, Dichloroaeetatε and rapamycin) on induction of rapid cell death by tetra-O-methy! nordihydroguaiaretic acid (M4N, EM1421, Teramcprocol) was studied in 4Tl and 67NR mouse breast cancer cell lines and LNCaP human prostate cancer cell line. M4N induced rapid TUNEL-positive cell death synergistically with either Rottlerin or Ly294002 in 4Tl and 67NR cell lines and with Rottlerin, Ly294002, Etoposide, or Dichloroacetate in LNCaP cells,
[087] MtN, Rottlerin, Ly2940()2, and Desferoxamine synergistically increased the translocation of phosphor-PKCδ (Thr05) into the nuclei. Transfeetion of PKCδ vectors enhanced cell death induced by M4N in both 4Tl and 67NR cells. On the other hand, transfeetion with dominant negative PKCδ (kinase negative) vectors partially suppressed cell death induced by a combination treatment of M4N with either Rottlerin or Ly294002.
Rottlerin but not Ly294002 depolarized mitochondrial membrane potential.
[088] The combination treatment of M4N with Ly294002 markedly suppressed tumor growth and metastasis in nude mice which had been orthotopically implanted with LNCaP tumors. By relieving tumor burdens, the drug combination has so far protected 100% of treated mice from death beyond 112 days while control mice all died before 52 days after tumor transplantation,
[089] Thus, chemicals which affect a very diverse range of cellular metabolisms that are often modulated specifically in cancer cells, such as the phosphatidyl inositol 3-kinase/Akt pathway (Ly294002), PKCδ/topoisomerase Ilα pathway (Ly294002, Etoposide, Rottlerin), or mitochondrial permeability transition (Rottlerin, Dichloroacetate) have the ability synergisticaily to enhance the tumoricidal effect of M4N on aggressive mouse and human cancers.
[090] M 4 N was evaluated in a mammary tumors at two different stages metastatic (cell line 4Tl) and nonmetastatic (cell line 67NR). M4N is effective to stop the growth of metastatic 4Tl cells but not sufficient to induce cell apoptosis within first 48 hours, although it is able to induce apoptosis in nonmetastatic 67NR cells, (20).
[091] A particularly compelling observation was that M4N together with low amounts of additional drugs (drugs that may be correlated with toxicity at higher does) is both efficacious and had essentially nontoxic side effects. In one method of the invention, M4N or another
NDGA derivative is administered daily with a metabolic inhibitor until an effective dosage is achieved in plasma and in the target cells to have the desired therapeutic effect without the toxicity associated with higher dosages that arc necessary when only one drug is administered. [092] Other features and advantages of the invention will be apparent from the detailed description, and from the claims.
DEFINITIONS:
[093] By "agent" is meant a polypeptide, peptide, nucleic acid molecule, small molecule, or mimetic.
[094] By "analog" is meant an agent having structural or functional homology to a reference agent.
|095] By "cell substrate" is meant the cellular or acellular material (e.g., extracellular matrix, polypeptides, peptides, or other molecular components) that is in contact with the cell,
[096] By "control" is meant a standard or reference condition.
[097] By "disease" is meant any condition or disorder that damages or interferes with the normal function of a cell, tissue, organ or subject,
[098] By "effective amount" is meant the amount of an agent required to ameliorate the symptoms of a disease relative to an untreated patient. The effective amount of an active therapeutic agent used to practice the present invention for the treatment of a disease or injury varies depending upon the manner of administration, the age, body weight, and general health of the subject. Ultimately, the attending clinician will decide the appropriate amount and dosage regimen.
[099] By "fragment" is meant a portion of a polypeptide that has at least 50% of the biological activity of the polypeptide from which it is derived. This portion contains, preferably, at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, or 90% of the entire length of the reference nucleic acid molecule or polypeptide. A fragment of a polypeptide or nucleic acid molecule may contain 10, 20, 30, 40, 50, 60, 70, 80, 90, or 100, 200, 300, 400, 500, 600,
700, 800, 900, or 1000 nucleotides or amino acids.
[0100] "HIF" refers to hypoxia inducible factor- 1
[0101] '"M4N" refers to Tetra-O-methyl nordihyroguaiaretic acid, EM 1421 or
Terameproeol, each of which are synonyms.
[0102] "Lower alkyl" and "lower alkoxy" refer to alkyl and alkoxy groups of 1-6 carbon atoms.
[0103] By "modifies" is meant alters. In the context: of the invention, an agent that modifies a cell, substrate, or cellular environment produces a biochemical alteration in a component (e.g., polypeptide, nucleotide, or molecular component) of the cell, substrate, or cellular environment.
[0104] "MTT" refers to 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide. [0105] " PDK-I" indicates 3'-phosphoinositide-dependent protein kinase-1. [0106] "PBfC" refers to phosphatidyl inosito 1-3 -kinase [0107] "mTOR" refers to the mammalian target of rapamycin.
[0108] As used herein, the terms "prevent," "preventing," "prevention," "prophylactic treatment" and the like refer to reducing the probability of developing a disorder or condition in a subject, who does not have, but is at risk of or susceptible to developing a disorder or condition.
[0109] As used herein, a ''prodrug'" is a pharmacologically inactive compound that is converted into a pharmacologically active agent by a metabolic transformation. [0110] By "subject" is meant a mammal, including, but not limited to, a human or non- human mammal, such as a bovine, equine, canine, ovine, or feline.
[0111] By "therapeutic delivery device" is meant any device that provides for the release of a therapeutic agent. Exemplary therapeutic delivery devices include osmotic pumps, indwelling catheters, and sustained-release biomaterials.
[0112] As used herein, the terms "treat," treating," "treatment," and the like refer to reducing or ameliorating a disorder and/or symptoms associated therewith. It will be appreciated that, although not precluded, treating a disorder or condition does not require that the disorder, condition or symptoms associated therewith be completely eliminated. [0113] By "variant" is meant an agent having structural homology to a reference agent but varying from the reference in its biological activity. Variants provided by the invention include optimized amino acid and nucleic acid sequences that are selected using the methods described herein as having one or more desirable characteristics. [0114] As used herein, "inhibiting" means slowing or stopping the growth of, [0115] As used herein, "with" or "along with" means that the compounds are administered during the same course of treatment, but not necessarily simultaneously. Administration may occur seconds, minutes, or hours apart in time, but will preferably be closely spaced (at least minutes). The compounds should be administered for sufficient duration (e.g. daily) so that an effective dosage is achieved in plasma and in the target tumor cells.
[Ot 16] Figure 1. Effect of M4N and/or desferoxamine on TUNEL-positive cell death and cell growth in 4Tl and 67NR cells. A and B: Either 4Tl (A) or 67NR (B) cells at subconfluent condition were treated with M4N (80μM) and/or desferoxamine (300μM). At 48hrs after treatment the cells were collected and TUNEL assay was performed. C; 1x10* 4Tl cells were seeded into 12 well plates. One day later, the cells were treated with different concentrations of MsN. The number of cells was measured by MTT assay at different times points. Data are presented as means (+/~) SD in triplicates.
proteins in PDK-1/Akt pathway. Either 4Tl or 67NR cells were seeded into 25mm2 flasks.
One day later, the cells were treated with M4 N (80μM) and/or Desferoxamine (75 or 150μM). Five hours later the cells were collected and the western blotting was performed for 4Tl ceils and 67NR cells, 0: without desferoxamine, 75: 75μM desferoxamine, 150: 150μM desferoxamine, (-): without M4N, and M4N: M4N (80μM). PDK-P: phospho-PDKl (Ser24i), Akt473P: phospho-Akt (Ser473), PRAS40P: phospho-PRAS40 (Thr246), RafP: phospho-Raf (Ser259), and MEK-P: phospho-MEKl/2 (Ser217'221).
TUNEL-positivε cell death in 4Tl and 67NR cells. A: Effect OfM4N, Desferoxamine. Rotllerin, Ly294002. and zVAD on TUNEL-positive cell death in 4Tl cells. 4Tl cells at subcontinent condition were treated with M4N (80μM), Desferoxamine (150μM), Rottlerin (5μM), or Ly294002 (20μM), At 24hrs after treatment the cells were collected and TUNEL assay was performed. When the effect of zVAD (50μM) was examined, zVAD had been added 30 minutes before other drugs were added. M; M4N, D: Desferoxamine, Ro: Rottlerin, Ly: Ly294002. B: Effect Of M4N5 Desferoxamine, Rottlerin, and Ly294002 on TUNEL- positive cell death in 67NR cells. 67NR cells at subconfluent condition were treated with M4N (80μM), Desferoxamine (150μM), Rottlerin (5μM), or Ly2940()2 (20μM). At 26hrs after treatment the cells were collected and TUNEI. assay was performed. Des: Desferoxamine, C: Effect of zVAD on TUNEL-positive cell death induced by M4N and Desferoxamine in 67NR cells, 67NR cells at subconfluent condition were treated with zVAD (50μM). and then treated with M4N (80μM) and/or Desferoxamine (150μM). At 42hrs later the cells were collected and TlJNEL assay was performed. Des: Desferoxamine, A-C; Data are presented as means (+/-) SD in triplicates. D: Effect Of M4N, Desferoxamine, Rottlerin. and Ly294002 on easpase-9 cleavage. Either 4Tl or 67NR cells were seeded into 25mm" flasks. One day later, the cells were treated with M4N (80μM), Desferoxamine (150μM). Rottlerin (5μM), and Ly294002 (20μM), Five hours later the cells were collected and the western blotting was performed for 4Tl cells and 67NR cells, IvI: IvI4N. 150: 150μM Desferoxamine, 150M: M4N plus 150μM Desferoxamine, Ro: Rottlerin, Ly: Ly294002. Full length caspase-9 is 5IkDa while cleaved caspase-9 is 39kDa. E&F: Effect of a combination treatment, of three drugs (M4N, Rottlerin, and Ly294002) together on TUNEL-positive cell death in 4Tl (E) and 67NR (F) cells. The concentration of M4N (M), Rottlerin (Ro), and Ly2940()2 (Ly) is 80μM, 5μM, and 20μM respectively. The combination treatment of Rottlerin and Ly2940Q2 is indicated by either Ro÷Ly or RoLy. Cell death was measured 19hrs after treatment. Data are presented as means (+/-) SD in triplicates. [0119] Figure 4. Effect Of M4N, Rottlerin, Ly294002, and Desferoxamine on the expression and cellular localization of total PKCδ and phosphor-PKCδ (Thr505) in 4Tl and 67NR cells 5hrs after treatment. A&B: The expression of the proteins in 4Tl (A) and 67NR (B) detected by the western blotting, The concentration OfM4N (M), Rottlerin (Ro)5 Ly294002 (Ly), and Desferoxamine (D) is 80μM, 5μM, 20μM, and 150μM respectively. The combination treatment of Desferoxamine and M4N is indicated by DM. Actin was used as control. C&D; The cellular localization of total PKCδ (C) and phosphor-PKCδ (Thr5'''5) in 4Tl cells. The concentration Of M4N, Rottlerin (Ro), Ly294002 (Ly), and Desferoxamine (Des) is 80μM, 5μM, 20μM, and 150μM respectively. Des (-) indicates 'without Desferoxamine treatment'. Des (+) indicates 'with Desferoxamine treatment'. [0120] Figure 5. Effect of PKCδ wild type and K376 mutant PKCδ (kinase negative) type vectors on TUNEL-positive cell death induced by M4N, Rottlerin, and Ly294002 in 4Tl (A) and 67NR (B) cells 24hrs after treatment. The concentration of M4N (M), Rottlerin (Ro), Ly294002 (Ly), and Desferoxamine (Des) is 80μM, 5μM, 20μM, and 150μM respectively. A (4Tl cells): The difference between the control and the PKCδ wild type in cells treated with M4N is statistically significant by t-test (5%). The difference between the control and the PfCCδ wild type in cells treated with M4N and Rottlerin is statistically significant by t-test (5%). The difference between the control and the PKCδ K376 mutant in cells treated with M4N and Rottlerin is statistically significant by t-test (5%). The difference between the control and the PKCδ in cells treated with M4N and Ly294002 is statistically significant by t- test (2%), B (67NR cells): The difference between the control and the PKCδ wild type in ceils treated with M4N is statistically significant by t-test (5%). The difference between the control and the PKCδ K376 mutant in cells treated with M4N and Rottlerin is statistically significant by t-test (5%). The difference between the control and the PKCδ K376 mutant in ceils treated with iVljN and L.y294002 is statistically significant by Rest (2%). The difference between the control and the PKCδ in cells treated with M4N and Ly294002 is statistically significant by t-test (1%). C: Effect of Genistein (50μM) on TUNEL-positive eel! death induced by the combination treatment of M4N (80μM) with Rottlerin (5μM) or Ly294002 (20μM) in 4Tl cells at 28hrs after treatment. "RoM' and 'LyM' designate the combination treatment of M4N with Rottlerin or Ly294002 respectively. Data are presented as means (+/-) SD in triplicates (A-C).
[0121] Figure 6. Effect of M4N and Rottlerin on the survival time of 4Tl (A) and 67NR (B) tumor-bearing mice. A: 5x104 4Tl cells were inoculated into fat pads of mammary glands in Balb/c female mice. The injections of either M4N (lmg/shot) and Rottlerin (lOOμg/shot) started 7 days after inoculation of tumors, Drug injections were performed three days a week. B: 5xlO4 67NR cells were inoculated into fat pads of mammary glands in Balb/c female mice. The injections of either M4N (lmg/shot) and Rottlerin (lOOμg/shot) started 8 days after inoculation of tumors. Drug injections were performed three days a week. In either 4Tl or 67NR tumor-bearing mice there were five to six mice in each group. The percentage of mice thai have died by the date after tumor inoculation was shown for each group, [0122] Figure 7. Effect of combination treatment Of M4N ami Eottlerin on Balb/e mice inoculated with a small number of 4Tl tumor cells. SxIO3 4Tl cells were inoculated into fat pads of mammary glands in Balb/c female mice. Effect of combination treatment of M4N (lmg/shot) and Rottlerin (lOOμg/shot) was examined. Drug injections started 3 days after inoculation of tumors. Drugs were administered six days a week. A: The percentage of mice that have died by the date after tumor inoculation was shown for each group. B: At 18 days after inoculation, the tumors were excised from the mice and their weights were measured. Data are presented as means (-*-/-) SD. The difference between the control and the combination treatment is statistically significant by Student's t-test (2%). C: At 18 days after inoculation, the lungs were excised from the mice and the clonogenicity assay was performed. Data are presented as means (+/-) SD. The difference between the control and the combination treatment is statistically significant by Student's t-test (2%). [0123] Figure 8. Effect of M4N, Rottlerin, and Ly294002 on TUNEL-positive cell death (A), cell growth (B), and cellular localization of total PKCδ (C) & phosphor-PKCδ (Thr507) (D) in LNCaP cells. The concentration of M4N (M), Roitlεrin (R), and Ly294002 (L), is 80μM, 5μM, and 20μM respectively. Cell death was measured at 28hrs after treatment (A), Cell growth assayed by the MTT method was measured at 24hrs after treatment (B). Data are presented as means (+/-) SD in triplicates (A & B). The cellular localization of PKCδ (C) and phosphor-PKCδ (Thr507) (D) was examined at 5hrs after treatment [0124] Figure 9. mice orifcotropicaily implanted with LNCaP tamers without treatment There are eight mice with arbitrary designations on the right side of the panels. The pictures were taken after 3 to 6 weeks after inoculation of tumors. The mice were daily injected with vehicle only. Vehicle injections started 8 days after implantation of tumors. Metastatic tumors are circled by red dots in the pictures,
[0125] Figure 10. Metastatic tumors which appeared on the ventral side of nude (nu/nu) mice orthotropicaily implanted with LNCaP tumors with a combination treatment Of M4N with Ly294002. There are eight mice with arbitrary designations on the right side of the panels. The pictures were taken after 3 to 6 weeks after inoculation of tumors. The mice were daily injected with M4N (Img/shot) and Ly294002 (lOOμg/shot). Drug injections started 8 days after implantation of tumors. Metastatic tumors are circled by red dots in the pictures. [0126] Figure 11, Effect of combination treatment Of M4N and Ly294002 on nude (nu/nu) mice orthotropicaily implanted with LNCaP tumors. LNCaP tumors were orthofropieaϊly implanted into the vicinity of prostate glawds m male nude mice. Effect of combination treatment Of M4N (Img/shot) and Ly294002 (lOOμg/shot) was examined. Drug injections started 8 days after implantation of tumors. Drugs were administered 7 days a week. A: The percentage of mice that have died by the date after tumor inoculation was shown for each group. B: Many tumor lesions appeared on the body surface of tumor-bearing mice in one to two weeks after tumor implantation. The size of these tumor lesions was estimated by the calculation described in Materials & Methods. Data are presented as means (+/-) SD. The difference between the control and the combination treatment at both 5 and 6 weeks after tumor implantation is statistically significant by Student's t-test (5%). C: The lungs (a & b) and the internal thoracic region facing the lungs (c & d) from the control mice that died of cancer at 7 to 8 weeks after tumor implantation. The lungs (e-g) from the treated mice that were killed at 1 1 weeks after tumor implantation. D: The tumors from the treated mice that were killed at 1 1 weeks after tumor transplantation. The tumors were cut by the scalpel so that their inside could be seen, a: the tumor in the prostate region, b-d: the tumors in the abdominal skin.
[0127] Figure 12. Effect of combination treatment of M4N asid Ly 294002 on metastatis tumors in the hmg and the skin in nude (nu/nu) mice orthotropicaily implanted with LNCaP tumors. A: Histological images of the lungs from eight different control mice that died of cancer at 7 to 8 weeks after tumor implantation (a-h). Metastasis lesions (designated as 1M') exist in six out of eight those mice. Notice that there is not any obvious metastasis lesion in the lung from all four different treated mice thai were killed at 1 1 weeks after tumor implantation (i-1). B; Many tumor lesions appeared on the body surface of tumor-bearing mice in one to two weeks after tumor implantation. Histological images of these lesions from either seven different control mice that died of cancer at 7 to 8 weeks after tumor implantation (a-g) or four different treated mice that were killed at 1 1 weeks after tumor implantation (i-1). All the sections were stained with Hematoxylin and Eosin.
appeared øis the ventral side of T8 mouse orthotropically inoculated with LNCaP cells assd treated with M4N and Ly294ΘO2. A: Images of metastatic tumors on the ventral side of T8 mouse from 6 to 22 weeks after inoculation of tumors. B: Total volumes of metastatic ventral tumors in T8 mouse from 3 to 22 weeks after inoculation of tumors. T8 mouse in this figure is the same one as T8 mouse in figure 3. The mice was daily treated with M4N (Img/shot) and Ly294Q02 (lOOμg/shot). Drug injections started 8 days after implantation of tumors. Rottlerin (lOOμg/shot) was additionally administered to the mice 10 weeks after the tumor inoculation.
[0129] Figure 14. A & B: Effect of the combination treatment Of M4N with different metabolic modulators on TUNEL-positive cell death in LNCaP cells with different concentrations of drags 24hrs or 48 brs after treatment. LNCaP cells were treated with M4N in the combination with either Rottlerin (A) or Ly294002 (B). Etoposide (C), Dichloroacetate (D), or Rapamycin (E&F) at either 24hrs (C5 D & E) or 48hrs (F). M4N (40μM, 60μM or 80μM), Etoposide (5, 10, or 20μM), Dichloroacetate (10 or 3OmM) or Rapamycin (5, 10, 20, or 30μM) were used. Data are presented as means (-*-/-) SD in triplicates (A-F).
[0130] Figure 15. Effect of combination treatment Of M4N with either Ly294002, Rottlerin plus Ly294002 or etoposide on nude (nu/nu) mice orthotropically implanted with LNCaP tumors. LNCaP tumors were orthotropically implanted into the vicinity of prostate giands isi male made mice. A; Effect of the combination treatment Of M4N with either Ly294002 or Rottlerin plus Ly294002 on the life-span of tumor-bearing mice. The percentage of mice that have died by the date after tumor inoculation was shown for each group. The numbers of mice in each group were 18, 5, 4, 8, and 9 for the control, Rottlerin alone, M4N alone, M4N & Ly294002, and M4N & Rottlerin & Ly294002 group respectively. Dosages of each injection were Img/shot (for M4N)5 0.2mg/shot (for Ly294002), and O.lmg/shot (for Rottlerin). Drug injections started 3 days after implantation of tumors. Drugs were administered 7 days a week for four weeks. After that drugs were injected once a week. B: Effect of the combination treatment Of M4N with either Etoposide or Rapamycin on the life-span of tumor-bearing mice. The percentage of mice that have died by the date after tumor inoculation was shown for each group. The numbers of mice in each group were 18, 5, 4, 4, 9 and 5 for the control, Etopside alone, Ly294002 alone, Rapamycin alone, M4N alone, M4N & Etoposide, and M4N & Rapamycin group respectively. Dosages of each injection were Img/shot (for M4N), 0.4mg/shot (for Etoposide), and 0.375mg/shot (for Rapamycin). Drug injections started 3 days after implantation of tumors. Drugs were administered 7 days a week for four weeks. After that drugs were injected once a week. C: Histological images of the lung from the tumor-bearing mice treated with different methods. Hematoxylin and Eosin staining. 10Ox magnification images. M indicates metastasis lesions. Cont, Eto, M4N, M4N -S-Ly5 M4N +Ro+Ly, and M4N +Eto indicate 'control', 'Etoposide alone', ' M4N alone', ' M4N and Ly294002 combination', ' M4N, Rottlerin, and Ly294002 combination', and ' M4N and Etoposide combination'.
[0131] Figϊire 16, Effect of eombsMtlϋE treatment of M4N with either Rottierin or Rapamycin in various tumor ceil lines. A: TUNEL-positive cell death in various tumor eel! lines treated with M4N and Rotterin for 24hrs, B: TUNEL-positive cell death in various tumor cell lines treated with M4N and Rapamycin. The concentration of M4N, Rottlerin, and Rapamycin is 80μM, either 10 or 20μM, and lOμM respectively. Data are presented as means (+/-) SD in triplicates.
DETAILED DESCRIPTION AND EXAMPLES [0132] Materials and Methods
[0133] Cell Culture Either cell line 4Tl or 67NR was a gift from Dr. Miller (20). The cells were cultured in RPMI 1640 medium supplemented with glucose (14mM), pyruvate (ImM), penicillin (100units/ml)5 streptomycin (10()μg/ml), and fetal bovine serum (10%), buffered with 25mM HEPES (pH 7.4). LNCaP human prostate cancer cell line was purchased from American Type Culture Collection (Manassas, VA). The cell line was cultured in RPMΪ1640 medium supplemented with glucose (14mM), pyruvate (ImM), penicillin (100units/ml), streptomycin (lOOμg/ml), and fetal bovine serum (10%). Drugs were dissolved in dimethyl sulfoxide (DMSOj ai a suitable concentration so that the final concentration of DMSO in the medium was less than 0.1 %, [0134] Reagents: M4N was synthesized and supplied by Erimos Pharmaceutical, L. L. C. (Raleigh, NC), according to the method described (87). Rottlerin was from Calbiochem (San Diego, CA). Ly294002 was from Cell Signaling Technology (Danvers, MA). zVAD-fmk was from R&D systems (Minneapolis, MN). Mouse anti-actin antibodies were from Sigma (Saint Louis, MO). Rabbit anti~HIF, anti-caspasε 9, anti-phospho-PDK! (Ser241), Akt, phospho-Akt (Ser473) (193H12), Phospho-Raf (Ser2S9), and Phospho-MEKl/2 (Ser217'7'1) antibodies were obtained from Cell Signaling Technology (Danvers, MA). Rabbit anti- phospho-PRAS40 (Thr246) antibody was from Biosource (Camarillo, CA). Desferoxamine mesylate and Etoposide were from Sigma (Saint Louis, MO). Sodium Dichloroacetate was from Alfa Aesar (Lancashire, UK). Mouse anti-Actin antibody was from Sigma (Saint Louis, MO). Rabbit anti-protein kinase Cδ (C-terminal) and anti-phosphor-protein kinase Cδ (Tyrl87) were from Santa Cruz Biotechnology (Santa Cruz, CA). Rabbit anti-caspase 9 and phospho-protein kinase Cδ (Thr505) antibodies were obtained from Cell Signaling Technology (Danvers, MA).
[0135] Animals; Female Balb/c mice were obtained from Charles River Laboratories (Wilmington, MA). M4N and Rottlerin were dissolved into a solvent, CPE (25/30) solvent system supplied by Erimos Pharmaceutical, L. L. C. (Raleigh, NC) and injected into mice either intravenously or intraperitoneally, T-cell deficient male nude mice (nu/nu) were obtained from Charles River Laboratories (Wilmington, MA). M4N and Ly294002 were dissolved into a solvent, CPE (25/30) solvent system supplied by Erimos Pharmaceutical, L.L.C. (Raleigh, NC) and injected into mice intravenously.
[0136] Tumor inoculation: Either 4Tl or 67NR cells were grown as described above. Cells growing subconfluently were collected and resuspended into the tissue culture medium minus fetal bovine serum and antibiotics, After counting the number of the cells, the cell concentration was adjusted. Twenty μl of the solution containing a selected number of cancer cells (either 5x103 or 5xlO4 cells) was injected into each fat pad of a pair of mammary glands located close to lower limbs. Tumor size was measured by weighing tumors extracted from mouse cadavers.
[0137] Clonogeπkity assay for luϊig metastasis: Clonogenicity assay was done based on the method by Pulanski & Ostrand- Rosenberg (80). At a selected time following tumor inoculation, mice were euthanized, mouse lungs were removed and minced well. Minced lungs were digested in 1 rag/ml collagenase type IV (Sigma, Saint Louis, MO) in phosphate buffered saline without magnesium and calcium (PBS (-)) at 37C for 2hrs. Digested lungs were filtered through 70μM cell strainers to remove debris. Cells were washed with PBS (-) and resuspended in the cell culture medium described above, supplemented with 60μM thioguanine (Sigma, Saint Louis, MO). After culturing the cells for several days, the numbers of emerging cell clones were counted,
[0138] MTT assay; Cells were incubated in PBS (-) supplemented with 5% fetal calf serum and G,5mg/ml MTT, 3-(4,5-Dimethy!thiazol-2-yi)-2,5-diphenyltetrazoliura bromide (Sigma, Saint Louis, MO) for 2hrs. After the incubation, the cells were dissolved in dimethyl sulfoxide and the color of the solutions was measured by a spectrophotometer at wavelengths of 540 and 690nm. The values correlated with viable cell numbers were obtained by subtracting the optical density at 690nm from that at 540nm.
[0139] Apoptosis assay: Simple TLINEL assay was conducted by using TlJNEL apoptosis detection kit (Upstate, Temecula, CA) with some modification. For the evaluation of the effect of PKC δ wild type and dominant negative vectors on TUNEL-positive cell death, we first transfected 4Tl or 67NR cells with these vectors by effectene (Qiagen, Valencia, CA). Wild type PKCδ vector is constructed from wild type PKCδ gene and pEGFP (Promega, Madison, WI). Dominant negative PKCδ vector is constructed from PKCδ gene mutated at a 376th amino acid residue (threonine) and pEGFP (Promega, Madison, WI) This threonine residue is located in ATP -binding domain of protein kinase Cδ, and therefore essential for any kinase reaction of the protein so that this mutant protein supposedly function as dominant negative. As a control we used pEGFP-N3 vector was used. These vectors contain green fluorescent protein (GFP) gene to be used as a transfection marker. The vectors are kind gifts from Dr. Yuspa (National Institutes of Health, Bethesda, MD) (88). Twenty four hours after transfection, the cells were treated with M4N, Rottlerin, or Ly294002. Then 24 hrs after this treatment with these reagents, the cells were fixed with 10% formaldehyde in phosphate buffered saline without calcium and magnesium (PBS (-)). The cells were then incubated with rabbit anti-GFP antibody (MBL International, Woburn, MA) followed by treatment with anti-rabbit IgG antibody conjugated with fluorescein (Promega, Madison, WI). TUNEL staining was conducted by using TUNEL apoptosis detection kit (Upstate, Temecula, CA) except for using Streptavidin-Alexa Fluor 568 (Molecular Probes, Eugene, OR) instead of Avidine-Rliodamine contained in the kit. Cell death was evaluated by dividing the number of TUNEL-positive cells among the cells positive with anti-GFP staining by the number of these GFP-posiiive cells. Western blotting; After cells had been grown in 25mm2 flasks and treated with reagents, the cells were washed with PBS (-) three times and suspended in RIPA buffer (15OmM NaCl, 5OmM Tris-HCl (pH 8,Q)5 0.1% SDS5 1% NP4O5 and 0.5% deoxycholate) supplemented with protease inhibitor cocktail (Calbiochem, San Diego, CA). The sample volumes were adjusted by the total protein amount. Protein assay was performed by Bio-Rad Protein Assay (Bio-Rad Laboratories, Inc. Hercules, CA). The samples were resolved by the standard SDS-polyacrylamide get electrophoresis and transferred to nitrocellulose membrane (Arnεrsham Bioscienees, Bjorkgatan, Sweden). The membranes were blocked with skim milk, and incubated with primary antibodies at 4°C overnight and then with secondary antibody conjugated with horse radish peroxidase at room temperature for 2hrs. The signals were detected by western blot chemiluminescence reagent plus (New England Nuclear Life Science Products, Boston, MA).
[0141] Surgical orthotropic implantation of LNCaP tumors: LNCaP cells were grown as described above. Cells growing subconfluently were collected and resuspended into the tissue culture medium without fetal bovine serum and antibiotics. After counting the number of the cells, the cell concentration was adjusted using the same medium. After 20 μl of the medium containing a selected number of cancer cells (about 5x107 cells) had been mixed together with the same volume of Matrigel (BD science, Bedford, MA), the combined solution was injected into the skin of nude mice. The tumor tissue growing subcutaneously was used for surgical orthotropic implantation of the tumor into nude mice, according to the method described by Wang et aL (89). The tumor tissue extracted from the skin was excised into pieces of about 2mm diameter, After nude mice were anesthetized by 2,2,2-Tribromoethanol (Aldrich Chemical Co. Inc., Milwaukee, Wf ), a small incision was made at the abdomen of each mouse and a tumor tissue piece was implanted in the neighbor of the prostate of each mouse. Eight days after the operation, the injection of drugs was initiated, and the drugs were injected intravenously every day for the indicated periods
[0142] ϊmmunoεytocheimstiy: Cells were cultured on glass cover slips coated with poly-L- ornithine (Sigma, Saint Louis, MO). At 5 hrs after treatment with drugs, the cells were fixed with 10% formaldehyde in diluted in PBS (-) and washed with PBS (~) three times. After permeabilized by 0.2% Tryton X-100 and 1% goat serum diluted in PBS (-), the cells were blocked by PBS (-) containing 5% goat serum. The cells were then incubated with primary antibodies diluted in PBS (-) containing 0.5% bovine serum albumin (BSA), and with secondary antibodies, which is either anti-Igϋ conjugated with fluorescein (Vector Laboratories. Buriingame, (LA), diluted in PBS (~) containing 0,5% BSA. The cells were observed through Zeiss fluorescent microscope (Carl Zeiss, Thomwood, NY). EXAMPLES
[0143] M4N has undergone Phase I/II clinical trials in patients by intravenous infusion. Two initial reports of these trials showed that M4N was able to cause long term stabilization of disease in some patients. However, tumor shrinkage has rarely been seen in patients following MφN Infusion as only limited amount of the drug can be delivered to tumors in situ by the method used .
[0144] To increase favorable clinical outcomes, several metabolic modulators have been administered in conjugation with M4N, resulting in rapid induction of tumor apoptosis and reduction of tumor metastases in both mouse breast cancer and human prostate cancer xenograft models. Example 1
[0145] Effect Of M4N to induce cell death in 4Tl and 67NR cells. 4Tl and 67NR cells are the cell lines derived from subpopulations of a mouse breast cancer (20). These two cell lines were characterized by their different behaviors regarding with metastasis. After we had injected either 4Tl or 67NR tumor cells into fai pads of mammary glands of Balb/c female mice, we measured the lung metastasis by the clonogenicity assay (80). At 4 weeks after injections, we did not detect clones in the lungs from 67NR cells while we detected many clones in the lungs from 4Tl cells in all the mice examined (data not shown). The data confirmed that 4Tl ceils used in this experiment were more metastatic than 67NR cells, as described earlier (20), Next we examined the effect of M4N, and desferoxamine maleate, a chemical known to mimic hypoxic conditions on cell death, by preventing HIF- lα degradation at Normoxia(21% O2) in 4Tl and 67NR cells (42,43). We measured cell death by TUNEL assay. At 48hr after treatment with M4N we detected more cell death in 67NR cells than 4Tl cells (Figure IA & I B). Desferoxamine (300μM) enhanced cell death induced by M4N in either 4Tl or 67NR cells, Since we did not detect many TLJNEL-positive cells in 4Tl cells, we measured the effect of MiN on cell growth in 4Tl cells by MTT assay (Figure 1C). MTT assay showed that M4N reduced the cell growth of 4Tl cells depending on the concentration of the drug. Even at 20μM of the drug, there was a significant effect of the drag to reduce cell growth, Cell growth stopped at 80μM, The effect of M4N on cell growth was evident at 42hrs after treatment. [0146] Effect of M4N on phosphorylation status of PDK-1/Akt pathway-related proteins in 4Tl and 67NR cells. Since we found differences between 4Tl and 67NR cells not only in metastasis capability but also susceptibility to M4N treatment, we next examined the effect of M4N and desferoxamine on the phosphorylation status of PDK-1/Akt kinase-related proteins of this pathway, which are known to be involved in cell growth and survival (44-47). Strikingly, we found that there were much less phosphorylated Raf and MEK-P detected in 4T] cells compared to that in 67 NR (Fig. 2). ϊn addition, we found that at 5hrs after treatment there was not much effect in 4Tl cells by either M4N or desferoxamine on the expression of phosphorylated PDK (Ser241), phosphorylated Akt (Ser473), phosphorylated PRAS40 (Thr246), phosphorylated Raf (Ser259), and phosphorylated MEK (Ser217/221) (Fig. 2). On the contrary, at 5hrs after treatment with M4N in 67NR cells there was significant reduction in the expression of phosphorylated PDK (Ser241), phosphorylated Akt (Ser473), phosphorylated PRAS40 (Thr246), phosphorylated Raf(Ser259), and phosphorylated MEK (Ser217/221) (Figure 2). PRAS40 (81, 82), Raf (83), and MEK (84) are all downstream phosphorylation targets of Akt. Total Akt expression was not changed much by M4N in either 4Tl or 67NR cells at 5hrs after treatment (Figure 2). Desferoxamine induced HΪF stabilization in both 4Tl and 67NR cells, as expected and desferoxamine at 150μM, a sensor of hypoxic condition, protects HIF very efficiently in both 4Tl and 67NR cells (39). Desferoxamine-mediated HIF induction was slightly suppressed by M4N in 4Tl cells while it was instead enhanced by M4N in 67NR cells (Figure 2). Overall the data indicates that M4N suppressed PD 1C-]. /Akt- related protein phosphorylation in 67NR cells while it failed to modulate it in 4Tl cells. Example 3
[0147] Effect of combination treatment Of M4N with PDK-I -related inhibitors, Ro or Ly on cell death in 4Tl and 67NR cells. Since we failed to quickly induce a substantial amount of TUNEL-positive cell death in 4Tl cells, we examined whether a combination treatment of M4N with other chemicals might be able to induce more cell death in 4Tl cells. For this attempt, we chose Ro and Ly since we observed some difference in the effect of M4N on PDK-1/Akt-related protein phosphorylation between 4Tl and 67NR cells (Fig, 2). Neither Ro nor Ly alone induced TUNEL-positive cell death in either 4Tl or 67NR cells (Figure 3). However, a combination treatment of M4N and Ly induced more cell death than M4N treatment alone in either 4Tl or 67NR cells (Figure 3A & 3B), Meanwhile Ro greatly enhanced cell death induced by M4N in 4Tl cells while it enhanced to some extent cell death induced by M4N in 67NR cells as well (Figure 3 A & 3B). Desferoxamine generally augmented ceil death even more, We examined the effect of pan-caspase inhibitor, zVAD, on the TUNEL-positive cell death if this cell death was caspase-dεpendent. The data showed that zVAD at 50μM didn't affect TUNEL-positive cell death in either 4Tl or 67NR cells (Figure 3A & 3C), Then we examined the easpase-1? cleavage, l he data shows that although a small amount of caspase-9 is activated by Ro In 67NR cells and this activation is slightly further augmented by M4N treatment, caspase-9 activation is overall very weak in either 4Tl or 67NR cells (Figure 3D). Overall the data suggest that rapid induction of 4Tl and 67NR cells by the combination drug treatment profoundly involves caspase-independent apoptosis mechanisms. We next examined if the combination treatment of M4N with both Ro and Ly induced more cell death than M4N with either Ro or Ly alone. The data indicates that Ro and Ly augments M4N-mediated cell death synergistically in 4Tl cells and additively in 67NR cells (Figure 3E & 3F), This supports the conclusion that M4N5 Ro, and Ly should be useful in combination for anti-cancer treatment. Example 4
[0148] Effect of M4N, Ro, Ly, and Des on the expression of total PKCδ and phosphor- PKCδ (Thr505) in 4Tl or 67NR cells. PKCδ has been known to be involved in certain types of cell death (27-29). To see if a synergistic induction of M4N -mediated cell death by Ro or Ly occurs through PKCδ, we examined the expression of PKCδ and its phosphorylated form at Thr505 residue in 4Tl and 67NR cells at 5hrs after treatment by the western blotting using phosphor-specific antibodies. Since TUNEL-positive cell death is already very evident in 24hrs after the combination treatment in this study, we focused on the modifications of PKCδ at 5hrs after the treatment, assuming that the event which causes cell death need to happen well before the final execution of cell death. Expression of total PKCδ protein was not affected much by the drug treatment alone or in different ways of combinations in both 411 cells and in 67NR cells, However the amount of phosphorylated PKCδ at Thr505 residue are significantly increased with M4N plus Ro and MsX plus Ly combinations in both 4Tl and 67NR cells, especially under conditions mimicking hypoxia through Desferoxamine treatment (Figure 4 A & 4B).
[0149] Cellular localization of total PKCδ and ssisekar translocation of phosphor-PKCδ
(Thr50*5) in 4TI cells after treatment with M4M, Ro, Ly, and Des. lhere were several reports suggesting that translocation of PKCδ into nuclei was required for induction of apoptosis (32, 33), ϊt was also shown that kinase negative full length PKCδ could not be induced to translocate into nuclei by apoptotic stimuli (32) Those un^nv^ seem to imply that phosphor-PKCδ localized in nuclei might be somehow involved in ihe mechanism of
J>.) apoptosis. As shown in Figure 4A and 4B, we found that M4N, Ro, Ly, and Des induced phosphorylation of PKCδ at Thr505 residue in both 4Tl and 67NR cells. To examine cellular distribution of both total PKCδ and phosphor-PKCδ (Thr505), we performed immunohistochemical staining in 4Tl cells after combination treatment of M4N, Ro, Ly, or Des for 5hrs (Figure 4C & 4D). We observed the total PKCδ is distributed more in cytoplasm than nuclei in 4Tl cells (Figure 4C). M4N, Ro, or Ly treatment alone did not change much the cellular distribution of total PKCδ in the cells. Des did not modulate much nuclear staining of total PKCδ either (Figure 4C). However, a combination treatment of M4N with either Ro or Ly in the presence of Des was found to facilitate the accumulation of phosphor- PKCδ (Thr505) in the nuclei in 4Tl cells (Fig. 4D). Example 6
[0150] Without any drug treatment, phosphor-PKCδ (Thr505) is distributed rather diffusively in 4Tl cells. M4N treatment augmented staining for phosphor-PKCδ (Thr505) in nuclei of the cells (Figure 4D). Ro also augmented staining for phosphor-PKCδ (Thr505) in nuclei. A combination treatment of M4N with Ro markedly augmented nuclear staining for phosphor- PKCδ (Thr505) in a very distinctive manner (Figure 4D). The nuclear membranous structure was very clearly visible under this condition. Although Ly treatment alone did not significantly modulate the staining in the cells, a combination treatment of M4N with Ly induced much more nuclear staining than M4N treatment alone. Des treatment alone only moderately increased nuclear staining in 4Tl cells. However, Des treatment in combination with M4N, M4N plus Ro, or M4N plus Ly markedly augmented nuclear staining in the cells (Figure 4D). Example 7
|0151] Direct evidence of PKCδ on induction of TUNEL-positive cell death by M4N, Ro, and Ly. The data described above indicate that M4N, Ro, and Ly modulate phosphorylation and cellular localization of PKCδ in both 4Tl and 67NR cells in less than 5hrs after treatment. We next examined if there is a causal relationship between the action of PKCδ and cell death. We transfected PKCδ or PKCδ dominant negative (PKCδK376) vectors and vector alone (control) into either 4Tl or 67NR cells and examined the impact of these vectors on cell death induced by the drug treatments in combination with M4N, Ro, and Ly. PKCδ dominant negative vectors are supposed to interfere in phosphorylation and nuclear translocation of PKCδ. As shown in Fig 5, neither the control nor PKCδ dominant negative vectors had much effect on cell death in both -4Tl and 67NR cells without drug treatment. However, transfection of PKCδ vectors induced cell death in 4Tl cells even without drug treatment (Figure 5A5 (-)).PKCδ vectors induced cell death significantly in 4Tl cells but not in 67NR cells while vector alone (control) or PKCδK.376 have little effect in induction of cell death in either 4Tl cells or 67NR cells (Figure 5A & 5B, (-)). Transfection of PKCδ vectors induced more cell death in both 4Tl and 67NR cells that were treated with M4N than transfection of control vectors (Figure 5A & 5B). Either Ro or Ly induced cell death synergistically with M4N in both 4Tl and 67NR cells that had been transfected with control vectors, in the same way as in the cells without transfection of any vectors (Figure 5A). Transfection of PKCδ vectors induced more cell death in both 4Tl and 67NR cells that were treated with M,jN plus Ro or M4N plus Ly than transfection of control vectors (Figure 5A & 5B). On the contrary, PKCδ dominant negative vectors (PKCδ K376) overall reduced the cell death induced by M4N, M4N plus Ro, or M4N plus Ly treatment in both 4Tl and 67NR cells (Figure 5A <ft 5B). We have also examined the effect of Genistein (50μM), known as the genera! Src tyrosine kinase inhibitor, on TUNEL-positive cell death induced by the combination treatment of M4N with either Ro or Ly in 4Tl cells (91). The data indicates that Genistein significantly suppressed the cell death (Figure 5C). Example 8
[0152] Effect of combination treatment of M4N with Ro on the life-span and tumor sizes in Balb/c mice inoculated with either 4Tl or 67NR cells. Lastly, we examined if the benefit of the combination treatment by M4N with Ro can be applicable to the in vivo study. Ro has been safely administered to mice by intranasal instillations in other studies. In this study, we used intravenous injection of drugs instead of intranasal instillations using CPE (25/30) solvent system for both M4N and Ro. We first inoculated 5x104 of either 4T1 or 67NR ceils into fat pads of mammary glands of Balb/c mice and examined if the drugs could elongate the life-span of these mice. We started to treat mice with drugs seven or eight days after the inoculation of 4Tl or 67NR cells respectively. We injected drugs three days a week after the initiation of drug treatment. Although either M4N or Ro alone didn't extend the lifespan of 4Tl -bearing mice, a combination treatment Of M4N with Ro extended the life span of these mice (Figure 6A). A similar tendency was found in mice inoculated with 67NR cells as well, As expected, mice overall survived longer with 67NR than 4Tl cell inoculations. While M4N or Ro alone didn't have much effect on the life-span of 67NR-bearing mice, a combination treatment of MjN with Ro overall extended the life span of the mice (Figure
Rxampk 9 [0153] Since 4Tl cells are very aggressive and kill mice in only two or three weeks period under this experimental condition, we examined the effect of a combination treatment by M4N and Ro on mice inoculated with a smaller number of 4Tl cells with more extensive chemotherapy protocol. In the next experiment we inoculated only 5x103 cells instead of 5x10' cells into fat pads of mammary glands of Balb/c mice. And also we started to inject the drugs three days after the inoculation of the cancer instead of seven days and we injected drugs six days a week instead of three days a week. Even with 5xlO3 cells per fat pad, 4Tl cells killed all the mice without treatment only in 2 to 5.5 weeks after the inoculation of the cells. On the contrary, the first mouse among the treatment group died almost 5 weeks after the inoculation of the cells. Overall the combination treatment extended the life-span of mice about 12 days (Figure 7A). The tumor size was compared at 18 days after the inoculation of the cancer between the control group and the treatment group. The data shows that the tumor size was significantly smaller in the treatment group than the control group (Figure 7B).
[0154] Effect of combination treatment of M4N with Ro on the metastasis to the lungs in Balb/c mice inoculated with 4Tl cells, measured by the donogemdty assay. The clonogeπicity assay was performed for the lungs from the mice either non-treated or treated with M4N plus Ro at 18 days after the inoculation of 5x103 4Tl cells into fat pads. The validity of this assay was confirmed by the experiment to compare metastatic ability between 4Tl and 67NR cells (data not shown). The data shows that the lung metastasis was extremely small in treated mice while lung metastasis was already very prevalent in the control mice (Figure 7C).
of M4N, Ro, and Ly on the cell death and growth and the cellular localization of total PKCδ and phosphor-FKCδ (Thr50') in LNCaF human prostate eøsseer cells, In the previous examples, we showed that Ro or Ly synergistically improved tumoricidal effect of M4N in mouse breast cancer cell lines, 4Tl and 67NR. Here we examined the effect of Ro and Ly on TUNEL -positive cell death induced by M4N in LNCaP human prostate cancer celts as an example for the application of the combination treatment to human cancer cells. As found in 4Tl and 67NR mouse breast cancer cells, either Ro or Ly synergistically augmented ceil death induced by M4N in LNCaP cells while Ro or Ly alone has no effect (Figure 8A) and cell death induced by M4N alone was also very small. On the other hand the MTT assay indicated that M4N treatment alone was able to reduce cell growth almost as much as a combination treatment of M4N with either Ro or Ly (Figure 8B). A single treatment with either Ro or Ly also suppressed cell growth to some extent. In three cancer cell lines that we have studied so far, our data clearly indicated thai although M4N alone was able to stop cancer cell growth, rapid cell death could be achieved only when M4N was used in combination with Ro and/or Ly (Figure 8A & B). We next examined the cellular localization of PKCδ and phosphor-PKCδ (Thr507) in LNCaP cells treated with M4N, Ro, and Ly for 5hrs. Neither M4N, Ro, nor Ly treatment caused any remarkable change in the expression of total PKCδ (Figure 8C) However, the combination treatment of M4N with either Ro or Ly induced substantial increase in total PKCδ in the nuclei (Figure 8C). The nuclear expression of phosphor-PKCδ (Thr507) was augmented by either Ro or M4N treatment (Figure 8D), A combination treatment of M4N with Ro induced more nuclear expression of phosphor-PKCδ (Thr507) than either Ro or M4N single treatment (Figure 8D). The extent of nuclear expression of PKCδ seems to be well correlated with the degree of the cell death under each condition (Figure 8A & 8C). Example 12
[0156] Effect of combination treatment of M4N with Ly on the life-span in nude (nu/nu) mice orthotropically implanted with LNCaP tumors. In the accompanied paper we already showed that Ro improved the anti-cancer effect of M4N on mouse breast cancer cells. Here we applied the synergistic induction of cell death by M4N and Ly observed under tissue culture condition to the animal experiments, using human prostate cancer cell line LNCaP. Ly was already safely administered to mice intraperitoneally before (60, 6). In this study we administered M4N and Ly dissolved into the solution by using CPE (25/30) solvent system to mice intravenously (87). LNCaP tumors were orthotropically implanted into the vicinity of the prostate of nude mice, We started to treat the mice with a combination treatment of M4N and Ly eight days after the implantation of LNCaP cells. Some tumors started to appear under the skin, especially in the abdomen, for both control and treated mice. While these tumors appeared under the skin all over the body in the control group, they appeared only in the lower abdomen area in the mice treated with the combination (Figure 9 & 10). All the mice injected with only vehicles died between 38 and 53 days after the tumor implantation. On the contrary, all the mice injected with M4N and Ly survived at 76 days after the tumor implantation (Figure HA). The size of these tumors increased very rapidly in the control mice while it increased very slowly in the treated mice (Figure 1 IB}. All the control mice died of cancer in 5 to 8 weeks alter tumor transplantation. Most of the mice from the control group already developed severe lung metastasis when they died (Figure HC, a-d). To compare the condition of the lung between the control and the treatment group, we killed some of the treated mice at 11 weeks after tumor transplantation. Unlike the lungs from the control group, those from the treatment group were devoid of obvious metastasis lesions (Figure 11C, e-g). We examined the tumor lesions in the prostate region and those on the skin in the abdomen area in the treated mice that were killed at 11 weeks after tumor implantation. The pictures show the inside of the tumors that had been cut by the scalpel (Figure HD). In most of the tumor lesions there were cavities inside. The region inside the tumor capsules was filled with white connective-tissue like materials. We examined the histology of metastasis lesions in the lungs and the skins (Figure 12). While we found very clear metastasis lesions in the lungs in 6 out of 8 control mice (Figure 12 A. a~h), we didn't find any metastasis lesions in the lungs of any mice treated with M4N and Ro (0 out of 4 mice) (Figure 12 A, i-1). While the tumors in the skins from the treated mice have large areas with lymphocyte infusions at their centers (Figure 12B, i-1), those from the control mice only have very small areas with lymphocyte infusions (Figure 12B, a-g).
[0157] The tumors under the abdominal skin in the mice treated with M4N and Ly were small in the first 6 weeks (Figure 10). However, the tumors increased in size after the first 6 weeks even with the treatment of M4N and Ly, We picked up one mouse (T8) to see if additional treatment with Ro might help to reduce the size of these tumors (Figure 13). The treatment with M4N, Ro, and Ly started at 9 weeks after the inoculation of tumors. Although the size of abdominal tumors was rapidly increasing between 6 and 9 weeks after the tumor inoculation, the tumors started to shrink very rapidly just after we changed the drug treatmeni from M4N and Ly combination to M4N, Ro, and Ly combination (Figure 13B). Example 14
[0158] Dosage dependent effect of M4N, Ro, and Ly on the cell death in LNCaP cells. We examined the effect of M4N, Ro, and Ly on cell death in LNCaP cells at many different concentrations of these drugs. This is a necessary step for applying this combination treatment to clinical usage. The data shows that M4N induced cell death synergistically with either Ro or Ly in LNCaP cells in a very broad range of concentrations of the drugs (Figure HA & B), Both Ro and Ly remained non-cytotoxic in tissue culture under this condition. The presence of either Ro or Ly made it possible for M4N to achieve great deal of cell death at kwer concentrations of K-LtN than in the absence of it, [0159] Effect of Etoposide, DkMoroacetate, &nά Rapamycm on TUNEL-positive cell death issdiseed by M4N in LNCaP cells. To explore other possible drugs to be combined with M4N5 we examined if Etoposide, Dichloroacetate, or Rapamycin can synεrgistically augment cell death with M4N (Figure 12. 15 & 16). We selected these drugs based on the speculations described in the introduction. The data indicates that either Etoposide, Diehloroacetate, or Rapamycin synergistically augmented TUNEL-positive cell death induced by M4N in 24hrs, as predicted from their action on cellular metabolism (Figure 14C, D, E & F), A combination treatment of M4N (40μM) with Rapamycin induced almost a hundred percent cell death in LNCaP cells at 48hrs after treatment (Figure 14F). This indicates that less than 40μM M4N would be enough under clinical conditions for the treatment to be effective. Example 16
[0160] Effect of combination treatment of M4N with either Ly, Ro plus Ly, Etoposide, or Rapamycin on the life-span and lung metastasis in nude (nu/nu) mice orthotropically implanted with LNCaP tumors. We applied the synergistic induction of cell death by M4N with either Ly, Ro plus Ly, Etoposide, or Rapamycin observed under tissue culture conditions to animal experiments (Figure 14 & 15). As in the previous experiment (Figures 9 to 13). in this study we administered M4N, Ly, Ro, Etoposide, or Rapamycin to mice intravenously using CPE (25/30) solvent system (87). LNCaP tumors were orthotropically implanted into the vicinity of the prostate of nude mice as in the previous studies (Figures 9 to 13), However, in this experiment we implanted about eight times the volume of tumors as in the previous experiments to detect the difference of efficacy of various combination treatments much faster. We started to treat the mice with combination treatments three days after the implantation instead of eight days, considering that the tumor volume is already large at the time of implantation in this experiment. This allows less time for possible metastasis to the transplanted tumors in this experiment than in the previous one. All the control mice died by 21 days after implantation of LNCaP tumors. All the mice treated with Etoposide alone died by 24 days after the implantation while all the mice treated with M4N alone died by 33 days, The combination treatment of M4N with Ly improved the survival of tumor-bearing mice although all the mice eventually died by 51 days (Figure 15A), This is an improvement compared with the survival time in the mice treated with either M4N or Ly alone. Three combination treatment of M4N, Ro, and Ly improved survival rate greatly. All the mice except lor one in this group (8 on! of 0) survived beyond 70 days after the implantation (Figure 15A). Meanwhile all the mice treated with M4N and either Etoposide (9 out of 9) or Rapamycin (5 out of 5) survived beyond 70 days after the implantation (Figure 15B). This is a great improvement compared with the survival time in the mice treated with either M4N, Etoposide, or Rapamycin alone.
[0161] Effect of the combination treatment on the lung metastasis (Figure ISC). The lungs of the control mice very often showed massive metastatic lesions (4 out of 5). On the contrary, the lungs from the mice treated with the combination treatment of M4N with either Ro plus Ly or Etoposide did not show any obvious metastasis at 80 days after the tumor implantation (0 out of 5 and 0 out of 3 respectively). The lungs from the mice treated with either Etoposide alone, M4N alone, or M4N plus Ly showed some metastasis lesions (3 out of 5, 2 out of 4, and 2 out of 8 respectively). The data clearly showed that all Ly, Ro, and Etoposide had capability to improve anti-cancer activity of M4N against LNCaP cells in animal experiments. Example 18
[0162] Effect of the combination treatment of M4N with either Ro or Rapamycin in various tissue culture cell lines. Lastly, we examined the combination effect of M4N with either Ro or Rapamycin in various tissue culture cells to see if these combination treatments are applicable to many tumor cells other than LNCaP cells. The data shows that the combination treatments effectively induce quick cell death in many tumor cell lines derived from various organs (Figure 16A & B).
[0163] From the foregoing description, it will be apparent that variations and modifications may be made to the invention described herein to adopt it to various usages and conditions. Such embodiments are also within the scope of the following claims. [0164] The recitation of a listing of elements in any definition of a variable herein includes definitions of that variable as any single element or combination (or subcombination) of listed elements. The recitation of an embodiment herein includes that embodiment as any single embodiment or in combination with any other embodiments or portions thereof.
[0165] All references, including patents, websites, programs, databases and publications, mentioned in this specification are herein incorporated by reference to the same extent as if each independent patent and publication was specifically and individually indicated to be incorporated by reference. 1. Megha, T., S, Lazzi, F. Ferrari, R. Vatti, CM. Howard, G. Cevenini, L, Leonicini, P. Luzi, Λ. Giordano, and P. Fosi. 1999, i expression of ihe G2-M checkpoint regulators cyclin Bl and P34CDC2 in breast cancer: a correlation with cellular kinetics. Anticancer Res. 19: 163-169,
2. Fang, F., and J. W. Newport. 1991. Evidence that the Gl-S and G2-M transitions are controlled by different cdc2 proteins in higher eukaryotes. Cell 66:731-742.
3. Dalton, S. 1992. Cell cycle regulation of the human cdc2 gene. The EMBO J. 11 :1797- 1804.
4. Morgan, D.O. 1995. Principles of cdk regulation. Nature 374:131-134.
5. Murray, A. W. 1992. Creative blocks: cell-cycle checkpoints and feedback controls. Nature 359: 599-604.
6. Kao, G.D., W.G. McKenna, and RJ. 1999, Muschel. P34(Cdc2) kinase activity is excluded from the nucleus during the radiation-induced G(2) arrest in HeLa cells. J. Biol, Chcm. 274:34779-34784.
7. Ambrosini, G., C. Adida, and D. C. Altieri. 1997. A novel anti-apoptosis gene, surviving, expressed in cancer and lymphoma. Nat. Med. 3:917-921 ,
8. Devcraux, Q, L.. and J.C. Reed. 1999. IAP family proteins-suppressors of apoptosis. Genes Dcv. 13:239-252.
9. O'Connor, D.S., D. Grossman, J. Plescia, F. Li, H. Zhang, A. Villa, S. Tognin, P.C. Marschisio, and D. C. Altieri. 2000. Regulation of apoptosis at cell division by p34cdc2 phosphorylation of surviving. Proc. Natl. Acad. Sci. U.S.A. 97:13103-13107.
10. Grossman, D., J. M. McNiff, F. Li, and D. C. Altieri. 1999. Expression and targeting of the apoptosis inhibitor, survivin, in human melanoma. J. Invest. Dermatol. 113:1076-1081.
1 1. Tamm, I., Y. Wang, E. SAusville, D.A. SCudiero, N. Vigna, T. Oltersdorf, and J.C. Reed. 1998. iAP-family protein surviving inhibits caspase activity and apoptosis induced by Fas (CD95), Bax, caspases, and anticancer drugs. Cancer Res. 58:5315-5320.
12. Hwu, J.R., W.N. Tseng, J. Gnabre, P. Giza, P, and R.C. Huang. 1998. Antiviral activities of methylated nordihydroguaiaretic acids. 1. Synthesis, structure identification, and inhibition of tat-regulated HIV transactivation. J. Med. Chem. 41 :2994-3000.
13. Gnabre, J.N., J.N. Brady, DJ. Clanton, Y. Ito, J. Dittmer, R.B. Bates, and R C. Huang. 1995. Inhibition of human immunodeficiency virus type 1 transcription and replication by DNA sequence-selective plant lignans. Proc. Natl. Acad, Sci. L.S.A. ^2: 1 1239-1 1243.
14. Chen, II., I . Feng, J.N. I i, R. Park, DAi, Mold, J. Gnabre, J.R. llwu, W.N. Tseng, and R.C, Huang. 1998. Antiviral activities of methylated nordihydroguaiaretic acids, 2. Targeting helpes simplex virus replication by the mutation insensitive transcription inhibitor tetra-O- methyl-NDGA. J. Med. Chem. 4l":3001-3007.
15. Craigo, J., M. Callahan, R.C. Huang, and A.L. DeLucia. 2000. Inhibition of human papillomavirus type 16 gene expression by nordihydroguaiaretic acid plant lignan derivatives. Antiviral Res. 47:19-28.'
16. Heller, J.D., J. Kuo, T.C. Wu, W.M. Kast, and R.C. Huang, 2001. Tetra-O-raelhyl nordihydroguaiaretic acid induces G2 arrest in mammary cells and exhibits tønoricidal activity in vivo. Cancer Res. 61 :5499-5504.
17. Chang, CC, J.D. Heller, J. Kuo, and R.C. Huang. 2004. Tetra-O-methyl nordihydroguaiaretic acid induces growth arrest and cellular apoptosis by inhibiting Cdc2 and surviving expression. Proc. Natl. Acad. Sci. U.S.A. 101 : 13239-13244.
18. Park, R., CC Chang, Y.C. Liang, Y. Chung, R.A. Henry, E. Lin, D.E. Mold, and R.C. Huang. 2005. Systemic treatment with tetra-O-methyl nordihydroguaiaretic acid suppresses the growth of human xenograft tumors. Clin. Cancer Res. 11 :4601-4609.
19. Yeh, H.-C, CM. Puleo, T.C Lim, Y.-P. Ho, P.E. Giza, R.C.C. Huang, and T.-H. Wang. 2006, A microfluidic-FCS platform for investigation on the dissociation of SpI-DNA complex by doxorubicin, Nucleic Acids Res. 34:el44.
20. Aslakson, CJ. , and F, R. Miller. 1992, Selective events in the metastatic process defined by analysis of the sequential dissemination of subpopulation of a mouse mammary tumor. Cancer Res: 52: 1399-1405.
21. Kikkawa, U.s Y. Takai, Y. Tanaka, R. Miyake, and Y, Nishizuka. 1983. Protein kinase C as a possible receptor protein of tumor-promoting phorbol esters. J. Biol. Chem. 258:11442- 1 1445.
22. KJkkawa, U,, A. Kishimoto. and Y. Nishizuka. 1989, The protein kinase C family; heterogeneity and its implications. Annu. Rev. Biochem. 58:31-44.
23. Jackson, D.N., and D.A. Foster, 2004. The enigmatic kinase Cδ: complex roles in cell proliferation and survival FASEB J. 18:627-636,
24. Brodie, C, and P.M. Blumberg. 2003. Regulation of cell apoptosis by protein kinase Cδ. tosis 8: 19-27.
25. Steinberg, S. F. 2004, Distinctive activation mechanisms and functions for protein kinase Cδ, Biochem. J. 384:449-459.
26. Watanabe, T.. Y. Ono, Y. Taniyama, K, Hazama, K. fgarashi, K. Ogita, U. Kikkawa, and
Y. Nishizuka. 1992. Cell division arrest induced by phorbol ester in CHO cells overexpressing protein kinase Cδ subspecies, Proc. Natl. Acad. Sci, U.S.A. 89: 10159-10163.
27. Acs, P., M, Beheshti, Z. Szallasi, L. Li, SJ-L Yuspa, and P.M. Blumberg. 2000. Effect of a tyrosine 155 to phenylalanine mutation of protein kinase Cδ on the proliferation and tumorigenic properties of NIH 3T3 fibroblasts. Carcinogenesis 21 : 887-891. 28. Stempka, L., A. Girod, H.-J. Muller, G, Rincke, F. Marks, M. Gschwendt, and D.
Bossemeyer. 1997. Phosphorylation of protein kinase Cδ (PKCδ) at Threonine 505 is not a prerequisite for enzymatic activity. J. Biol Chem. 272:6805-6811,
29. Stempka, L., M. Schnolzer, S. Radke, G. Rincke, F. Marks, and M. Gschwendt. 1999. Requirements of protinβ kinase Cδ for catalytic function. J. Biol. Chem. 274:8866-8892,
30. LIu5 Y., C. Graham, A. Li, RJ. Fisher, and S, Shaw, 2002. Phosphorylation of the protein kinase C-theta activation loop and hydrophobic motif regulates its kinase activity, but only activation loop phosphorylation is critical to in vivo nucSear-facor KB induction. Biochem, J. 361 :255-265.
31. Liu. Y., N.V. Belkina, C. Graham, and S. Shaw. 2006. Independence of protein kinase C- δ activity from activation loop phosphorylation. Structural basis and altered functions in cells. J. Biol. Chem. 281 :12102-12111.
32. DeVries, T. A., M.C. Neville, and M. E. Reyland. 2002. Nuclear import of PKCδ is required for apoptosis: identification of a novel nuclear import sequence. The EMBO J. 21 :6050-6060,
33. DeVries-Seiraon, T. A., A.M. Ohm, MJ, Humphries, and M. E. Reyland. 2007, Induction of apoptosis is driven by nuclear retention of protein kinase C delta. J, Biol, Chem. 282:22307-22314.
34. Emoto, Y., Y. Manome, G. Meinhardt, H. Kisaki, S. Kharbanda, M. Robertson, T. Gharyur, W. W. Wong, R. Kamen, R. Weichselbaum, and D. Kufe. 1995. Proteolytic activation of protein kinase C δ by an ICE-like protease in apoptotic cells. The EMBO J. 14:6148-6156.
35. Ghayur, T. M. Hugunin. R.V. Talanian, S, Ratnofsky, C, Quinlan, Y, Emoto, P. Pandey, R. Datta, Y. Huang, S. Kharbanda, H. Alien, R. Kamen* W. Wong, and D. Kufe. 1996. Proteolytic activation of protein kinase C delta by an 1CE/CED 3-Sike protease induces characteristics of apoptosis. J, Exp. Med, 184:2399-2404.
36. Blass, M,, L Kronfeld, G. Kazimirsky, P.M. Blumberg, and C. Brodie, 2002. Tyrosine phosphorylation of protein kinase Cδ is essential for its apoptotic effect in response to Etoposide. MoI. Cell. Biol. 22:182-195.
37. Yoshida, K., T. Yamaguchi, HL Shinagawa, N. Taira, K.I. Nakayama, and Y. Miki. 2006. Protein kinase C δ activates topoisomerase Ilα to induce apoptotic cell death in response to DNA damage. MoL Cell. Biol. 26:3414-3431,
38. Yang, J., S.A. Mani, J.L. Donaher, S. Ramaswamy, R. A. Itzykson, C. Come, P. Savaqner, I. Gitelman, A. Richardson, and R.A. Weinberg. 2004. Twist, a master regulator of morphogenesis, plays an essential role in tumor metastasis. Cell 117: 927-939.
39. Gordan, J.D,, and M.C, Simon, 2007 Hypoxia-inducible factors: central regulators of the tumor phenotype. Curr. Opin, Genet. Dev. 17:71 -77. 40. Wouters, A., B. Pauwels, F. Lardon, F, and J. B. Vermorken, 2007. Review; implications of in vitro research on the effect of radiotherapy and chemotherapy under hypoxic conditions. Oncologists 12:690-712,
41. Melilio, G. 2007. Targeting hypoxia cell signaling for cancer therapy. 2007. Cancer
Metastasis Rev. 26: 341-352. ~
42. Aldinucci, C5 A. Carretta, L. Ciccoli, S. Leoncini, C. Signorini, G. Buonocore, and G. P, Pessina. 2007, Hypoxia affects the physiological behavior of rat cortical synaptosomes. Free Radic. Biol. Med. 42:1749-1756.
43. Clavijo. C, J.-L. Chen, K.-J. Kim, M.E. Reyland, and D.K. Arm. 2007. Protein kinase Cδ-dependent and -independent signaling in genotoxic response to treatment of desferoxamine, a hypoxia-mimetic agent. Am. J. Physiol. Cell Physiol. 292:C2150-C2160.
44. Serova, M, A. Ghoul, K.A. Benhadji, E. Cvitkovic, S. Faivre, F. Calvo, F. Lokiec, and E. Raymond. 2006. Preclinical and clinical development of novel agents that target the protein kinase C family. Seminars in Oncology 33:466-478.
45. Da Rocha, A. B., D. R. Mans, A. Regner, and G. Schwartsmann. 2002. Targeting protein kinase C: New therapeutic opportunities against high-grade malignant gliomas? The Oncologist ?: 17-33.
46. Morgεnsziern, D., and H.L. McLeod. 2005. PDK/Akt/tnϊOR pathway as a target for cancer therapy. Anticancer Drugs. 16:797-803.
47. Faivre, S,, S. Djelloul, and E. Raymond. 2006. New paradigms in anticancer therapy: targeting multiple signaling pathways with kinase inhibitors. Semin. Oncol, 33:407-420.
48. Cully, M5 H. You, A.J. Levine, and T.M. Mak. 2006. Beyond PTEN mutations: the PI3K pathway as an integrator of multiple inputs during tuniorigersesis. Nature Rev. Cancer, 6: 184-
49. Osaki, 3VL5 M. Oshimura, and H. Ko. 2004. P!3K~Akt pathway :ϊts function and alterations in human cancer. Apoptosis, 9:667-676.
50. Manning, B, D., and L.C. Cantley. 2007. Akt/PkB signaling: Navigating downstream. Cell
129:1261 -1274.
51. Gschwendt, M., HJ. Mullεr, K. Kielbassa, R. Zang, W. Kittstein, G. Rincke, and F. Marks. 1994. Rottlerin, a novel protein kinase inhibitor. Biochem. Biophys, Res. Comm, 199:93-98.
52. Davies, S. P., H. Reddy, IvS. Caivano, and P. Cohen. 2000. Specificity and mechanism of action of some commonly used protein kinase inhibitors. Biochem, J. 351 (Pt l ):95-105,
53. Le Good, j.A.s W.H. Ziegier, D.B. Parεkh5 D.R. Alessi, P. Cohen, and PJ. Parker. 1998, Protein kinase C isotypes controlled by phosphoinositide 3 -kinase through the protein kinase PDKl . Science 281 :2042-
54, Jackson, D.N., and D. A. Foster. 2004. The enigmatic kinase Cδ: complex roles in cell proliferation and survival FASEB J. 18:627-636. 55. Brodie, C, and P.M. BIuraberg. 2003, Regulation of cell apoplosis by protein kinase Cδ. Apoptosis 8: 19-27.
56. Steinberg, S, F, 2004. Distinctive activation mechanisms and functions for protein kinase Cδ. Biochem. J. 384:449-459,
57. Tillman, D. M., K. Izeradjene, K.S. Szυcs, L. Douglas, and J. A. Houghton. 2003, Rottlerin sensitizes colon carcinoma cells to tumor necrosis factor-related apoptosis-inducing ligand- induced apoptosis via uncoupling of the mitochondria independent of protein kinase C. Cancer Res. 63:5118-5125.
58. Kim, E.-H., S.-U. Kim, and K.-S. Choi. 2005. Rottlerin sensitized gloma cells to TRAIL- induced apoptosis by inhibition of Cdc-2 and the subsequent downregulation of surviving and XIAP. Oncogene 24:838-849.
59. Ringshausen, L, M. Oelsner, K. Weick, C. Bogner, C. Peschel, and T. Decker. 2006. Mechanisms of apoptosis-induction by rottlerin: therapeutic implications for B-CLL. Leukemia 20:514-520.
60. Hu, L., C, Zaloudek, G..B. Mills, J. Gray, and R.B, Jaffe. 2000. In vivo and in vitro ovarian carcinoma growth inhibition by a phosphatidylinosito! 3-kinase inhibitor (LY294002). Clin. Cancer Res. 6:880-886.
61. Semba, S., N. Itoh, M. Ito, M. Harada, and M. Yaraakawa. 2002, The in vitro and in vivo effects of 2-(2-morpho!inyl)-8-phenyl-chromone (LY294002), a specific inhibition of phosphatidylinositol 3 '-kinase, in human colon cancer ceils. Clin. Cancer Res. 8:1957-1963.
62. Berrie, CP. 2001. Phosphoinisitide 3-kinase inhibition in cancer treatment. Expert Opin. Investg. Drugs, 10: 1085-98.
63. Stein, R.C. 2001 , Prospects for phosphoinositide 3-kinase inhibition as a cancer treatment. Endoer. Reat, Cancer. 8: 237-48)
64. Eckhardt, B. L., B. S. Parker, R.K. van Laar, CM. Restall, A.L. Natoli, M.D. Tavaria, K. L. Stanley, E. K. Sloan, J. M. Moseley, and R.L. Anderson. 2005. Genomic analysis of a spontaneous model of breast cancer metastasis to bone reveals a role for the extracelluar matrix. MoI. Cancer Res. 3:1-13.
65. Gordan, J.D., and M. C. Simon. 2007 Hypoxia-inducible factors: central regulators of the tumor phenotype. Curr. Opin. Genet. Dev. 17:71-77.
66. Manka, D., Z, Spicεr, and D. E. Millhorn. 2005. Bcl-2/adenovirus ElB 19kDa interacting protein-3 knockdown enables growth of breast cancer metastases in the lung, liver, and bone. Cancer Res. 15: 11689-1 1693.
67. Bonnet, S,, S. L. Archer, J. Al [alums-Turner, A. Barorny, C. BeauJieυ, R.. 1 hompson, CT. Lee,, GJ), Lopaschuk, L. Puttagimta, S. Bonnet, G, Harry, K. Hashimoto, CJ. Porter, K-I. A. Afsdrade, B. Thebaud, and E. D, Michelakis. 2007. Λ πiUochondπa-IC channel axis is suppressed in cancer and its normalization promotes apoptosis and inhibits cancer growth. Cancer Ceil 1 1 :37-51 , 68. Kroemer, G. & SJ. Martin. 2005. Caspase-indεpendent cell death. Nature Med. 11 :725- 730.
69. Shizukuda, Y., A, HeSisch, R. Yokota, and IA. Ware. 1999. Downregulation of protein kinase Cδ activity enhances endothelial ceil adaptation to hypoxia. Circulation 100:1909- 1916.
70. Baek, S.-H., U.-Y. Lee, E.-M Park, M.-Y. Man, Y.-S., Lee, and Y.-M. Park. 2001 , Role of protein kinase Cd in transmitting hypoxia signal to HSF and HIF-I. J, Cell. Phys, 188:223- 235.
71. Contβ, M., B. Soper, Q. Chang, and B. Tepperman. 2004. The role of PKCδ and PKCε in the neonatal rat colon in response to hypoxia challenge. Pediatric Res, 55:27-33.
72. Parekh, D. B., W. Ziegler, and PJ. Parker. 2000. Multiple pathways control protein kinase C phosphorylation. EMBO J. 19:496-503.
73. Newton, A.C. 2003. Regulation of the ABC kinases by phosphorylation; protein kinase C as a paradigm. Biochem J. 370 (Pt 2):361-371.
74. Rybin, V.O., A. Sabri, J. Short, J.C. Braz, J.D. Molkentin, and S.F. Steinberg. 2003. Cross-regulation of novel protein kinase C (PKC) isoform function in cardiomyocytes. J, Biol. Chem. 278:14555-14564.
75. Fukunaga, M., M. Oka, M. Ichihashi, T. Yamamoto, H. Matsuzaki, and U. Kikkawa. 2001. UV-induced Tyrosine phosphorylation of PKCδ and promotion of apoptosis in the HaCaT cell line. Biochem. Biophys. Res. Comm. 289:573-579.
76. Parekh, D.B., R..M. Katso, N.R. Leslie, CP. Downes, KJ. Procyk, MD. Waterfield, and
PJ. Parker, 2000. β l-Integrin and PTEN control the phosphorylation of protein kinase C, Biochem. J, 352:425-433,'
77. Kroemer, G., G. Lorenzo, and C. Brenner. 2007. Mitochondrial membrane permeabilization in cell death, Physiol. Rev. 87:99-163.
78. Parcel iier, A,s L. A, Tintignac, E, Zhuravleva, and B.A. Hemmings. 2008. PKB and the mitochondria: AKTing on apoptosis. Cellular Signal. 20:21-30.
79. Gupta, A. K., GJ. Cerniglia, R. Mick, M.S. Ahmed, V.J. Bakanauskas, RJ. Muschel, and W.G. Mckenna. 2003. Radiation sensitization of human cancer cells in vivo by inhibiting the activity of PI3K using Ly294002. Int. J. Radiation Oncol Biol. Phys. 56:846-853.
80. Pulaski, B.A., and S, Ostrand-Rosenberg. 1998. Reduction of established spontaneous mammary carcinoma metastasis following immunotherapy with major histocompatibility complex class II and B7.1. Cancer Res. 58: 1486-1493.
81. Vander Haar, E., S.I. Lee, S. Bandhakavi, TJ. Griffin, and D. H. Kim. 2007, Insulin signaling to mTOR mediated by the Akt/PKB substrate PRAS40. Nature Cell Biol, 9:316- 323.
82. Mackintosh, C, 2004. Dynamic interactions between 14-3-3 proteins and phosphoproteins regulate diverse cellular processes. Biochem, J. 381 :329-342. 83. Reuseh, H. P., S, Zimmermann, M. Schaefer, M Paul, and K. Moelling. 2001. Regulation of Raf by Akt controls growth and differentiation in vascular smooth muscle cells. J. Bioi. Chem. 276:33630-33637.
84. Chang, F., L.S. Steelman, J.T. Lee, J.G. Shelton, P.M. Navolanic, W.L. Blalock, R.A. Franklin, and J. A. McCubrey. 2003. Signal transduction mediated by the Ras/Raf/MEK/ERK pathway from cytokine receptors to transcription factors: potential targeting for therapeutic intervention. Leukemia 17:1263-1293.
85. Siddharth, K., V. Anautharam, Y. Yang, CJ. Choi, A. Kauthasamy, and A.G. Kauthasamy. 2005. Tyrosine phosphorylation regulated the proteolytic activation of protein kinase Cδ in dopaminergic neuronal cells. J. Biol. Chem. 280:28721-28730.
86. Li5 T.K. and L.F, Liu. 2001 , Tumor cell death induced by topoisomerase-targeting drugs.
Ann, Rev. Pharmacol. Toxicol. 41 :53-77.
87. Lopez, R.A., A.B. Goodman, M. Rhodes, J.A. Blomberg, and J. Heller. 2007. The anticancer activity of the transcription inhibitor terameproeol (meso~tetra-Q~methyl nordihydroguaiaretic acid) formulated for systemic administration. Anti-Cancer Drugs 18:933-939.
88. Li, L,, P.S. Lorenzo, K. Bogi, P.M. Blumberg, and S.H. Yuspa. 1999, Protein kinase Cδ targets mitochondria, alters mitochondrial membrane potential, and induces apoptosis in normal and neoplastic keratiocytes when overexpressed by an adenovirus vector. Mole. Cell. Biol 19:8547-8558.
89. Wang, X., Z, ZiIi, J, Geller, and R.M. Hoffman. 1999. High-malignancy orthotopic nude mouse model of human prostate cancer LNCaP. The Prostate 39: 182-186.
90. Chang CC, Liang YC5 Klutz A5 Hsu CI5 Lin CF5 Mold DE, Chou TC, Lee YC5 Huang RC. 2006. Reversal of multidrug resistance by two nordihydroguaiaretic acid derivatives, M4N and maltose-M3N, and their use in combination with doxorubicin or paclitaxel. Cancer Chemotherapy Pharmacol 58:640-653.
91. Kaul S, Anantharam V, Yang Y, Choi CJ, Kanthasamy A, and Kanthasamy AG. 2005 Tyrosine Phosphorylation Regulates the Proteolytic Activation of Protein Kinase C in Dopaminergic Neuronal Ceils. J. Biol. Chem. 280:28721-30

Claims

WE CLAIM:
1. A pharmaceutical composition comprising an effective amount of nordihydroguaiaretic acid (NDGA) or a derivative thereof of formula 1:
Figure imgf000050_0001
wherein R1, R2, R3 and R4 independently represent hydroxy, a straight or branched chain lower alkyl or alkoxy, an amino acid residue, a substituted amino acid residue, a nitrogen- containing 5- or 6-membered heterocyclic ring or a saccharide residue; the amino acid residue, substituted amino acid residue, nitrogen-containing 5 or 6 membered heterocyclic ring or saccharide residue being optionally joined to the phenyl ring by a linker of an oxygen atom and 1-10 carbon atoms; and an effective amount of a metabolic modulator.
2. The pharmaceutical composition of claim 1 comprising tetra-omethyl nordihydroguaiaretic acid (M4N) or maltose-M3N.
3. The pharmaceutical composition of claim 1 or 2 wherein the metabolic modulator is selected from the group consisting of an inhibitor of the PDK-I /PO K/ AKT pathway, an inhibitor of the PKCδ/topoisomerase Ilα pathway, an inhibitor of mitochondrial permeability transition, and an inhibitor of AKT/mTOR pathway.
4. The pharmaceutical composition of claim 3, wherein the metabolic inhibitor is selected from the group consisting of Ly294002, etoposide, rottlerin, dichloroacetate, cetuximab, trastuzumab, bevacizumab and rapamycin.
5. The pharmaceutical composition of claim 3 wherein the metabolic inhibitor is selected from the group consisting of etoposide, Ly294002, rapamycin, rottlerin,and water soluble- derivatives thereof.
6. A method of treating a tumor, comprising administering to a mammal in need of treatment an effective amount of the pharmaceutical composition of one of claims 1-5,
7. The method of claim 6 wherein the mammal is a human, cat, dog or mouse.
8. The method of one of claims 6-7 wherein the tumor is a solid tumor.
9. The method of one of claims 6-7 wherein the tumor is a hematological tumor.
10. The method of one of claims 6-8 wherein the tumor is malignant.
11. The method of claim 10 wherein the tumor is selected from the group consisting of breast cancer, prostate cancer, lung cancer, colon cancer and ovarian cancer.
12. A method of preventing or inhibiting tumor growth in an animal, said method comprising administering an effective amount of nordihydroguaiaretic acid (NDGA) or a derivative thereof of formula I:
Figure imgf000051_0001
wherein R1, R2, R3 and R4 independently represent hydroxy, a straight or branched chain lower alkyl or alkoxy, an amino acid residue, a substituted amino acid residue or a saccharide residue; the amino acid residue, substituted amino acid residue or saccharide residue being optionally joined to the phenyl ring by a linker of an oxygen atom and 1-10 carbon atoms; and an effective amount of a metabolic modulator.
13. The method of claim 12 wherein the NDGA derivative is tetra-o-methyl nordihydiOguaiaretic acid (M4N) or maltose-MβN.
14. The method of claim 12 or 13 wherein the mammal is a human, cat, dog or mouse.
15. The method of one of claims 12-14 wherein the metabolic inhibitor is selected from the group consisting of an inhibitor of the PDK-1/PI3K/AKT pathway, an inhibitor of the PKCδ/topoisomerase Ilα pathway, an inhibitor of mitochondrial permeability transition, and an inhibitor of the mitogen-activated protein kinase (MAPK/RAS) pathway.
16. The method of claim 15 wherein the metabolic inhibitor is selected from the group consisting of Ly294002, etoposide, Rottlerin, dichloroacetate, cetuximab, trastuzumab, bevacizumab and rapamycin.
17. The method of claim 15 wherein the metabolic inhibitor is selected from the group consisting of etoposide, rapamycin, and water soluble derivatives thereof.
18. The method of one of claims 12-17 wherein the tumor is a solid turner.
19. The method of one of claims 12-17 wherein the tumor is a hematological tumor.
20. The method of one of claims 12-19 wherein the tumor is malignant.
21. The method of claim 20 wherein the tumor is selected from the group consisting of breast cancer, prostate cancer, lung cancer, colon cancer and ovarian cancer.
22. A method of preventing tumor metastasis in a mammal, said method comprising administering an effective amount of nordihydroguaiaretic acid (NDGA) or a derivative thereof of formula 1 :
Figure imgf000052_0001
wherein R;, R2, R3 and R4 independently represent hydroxy, a straight or branched chain lower alky] or alkoxy, an amino acid residue, a substituted amino acid residue or a saccharide residue; the amino acid residue, substituted amino acid residue or saccharide residue being optionally joined to the phenyl ring by a linker of an oxygen atom and 1-10 carbon atoms; and an effective amount of a metabolic modulator.
23. The method of claim 22 wherein the NDGA derivative is tetra~β~methyl nordihydroguaiaretic acid (M4N) or maltose-M3N.
24. The method of claim 22 or 23 wherein the mammal is a human, cat, dog or mouse.
25. The method of one of claims 22-24 wherein the metabolic inhibitor is selected from the group consisting of an inhibitor of the PDK-I /PD K/A KT pathway, an inhibitor of the PK.Cδ/topoisomerase Ilα pathway, an inhibitor of mitochondrial permeability transition, and an inhibitor of the mitogen-activated protein kinase (MAPK/RAS) pathway.
26. The method of claim 25 wherein the metabolic inhibitor is selected from the group consisting of Ly294002, etoposide, Rottlerin, dichloroacetate, cetuximab, trastuzumab, bevacizumab and rapamycin.
27. The method of claim 25 wherein the metabolic inhibitor is selected from the group consisting of etoposide, rapamyein, and water soluble derivatives thereof.
28. The method of one of claims 22-27 wherein the tumor is a solid tumor.
29. The method of one of claims 22-27 wherein the tumor is a hematological tumor.
30. The method of one of claims 22-29 wherein the tumor is malignant.
31. The method of claim 30 wherein the tumor is selected from the group consisting of breast cancer, prostate cancer, lung cancer, colon cancer, ovarian cancer, brain cancer and pancreatic cancer.
32. A method of prolonging the life of a mammal having a malignant tumor, said method comprising administering to the mammal an effective amount of nordihydroguaiaretic acid (NDGA) or a derivative thereof of formula I:
Figure imgf000054_0001
wherein Ri, R2, R3 and R4 independently represent hydroxy, a straight or branched chain lower alkyl or alkoxy, an amino acid residue, a substituted amino acid residue or a saccharide residue; the amino acid residue, substituted amino acid residue or saccharide residue being optionally joined to the phenyl ring by a linker of an oxygen atom and 1-10 carbon atoms; and an effective amount of a metabolic modulator.
33. The method of claim 32 wherein the NDGA derivative is tetra-o-methyl nordihydroguaiaretic acid (M4N) or maltose-M3N.
34. The method of claim 32 or 33 wherein the mammal is a human, cat, dog or mouse.
35. The method of one of claims 32-34 wherein the metabolic inhibitor is selected from the group consisting of an inhibitor of the PDK-I /PI 3 K/ AKT pathway, an inhibitor of the PKCδ/topoisomerase Ilα pathway, an inhibitor of mitochondrial permeability transition, and an inhibitor of the mitogen-activated protein kinase (MAPK/RAS) pathway.
36. The method of claim 35 wherein the metabolic inhibitor is selected from the group consisting of Ly294002, etoposide, Rottlerin, dichloroacetate, cetuximab, trastuzumab, bevacizumab and rapamycin.
37. The method of claim 35 wherein the metabolic inhibitor is selected from the group consisting of etoposide, raparaycin, and water soluble derivatives thereof.
38. The method of one of claims 32-37 wherein the tumor is a solid tumor.
39. The method of one of claims 32-37 wherein the tumor is a hematological tumor,
40. The method of claim 38 wherein the tumor is selected from the group consisting of breast cancer, prostate cancer, lung cancer, colon cancer, ovarian cancer, brain tumor and pancreatic cancer.
41. A method of treating a tumor and/or prolonging survival time of a mammal afflicted with a tumor, said method comprising administering a compound of formula I:
Figure imgf000055_0001
wherein Rj, R2, R3 and R4 independently represent hydroxy, a straight or branched chain lower alkyl or alkoxy, an amino acid residue, a substituted amino acid residue, a nitrogen- containing 5- or 6-membered heterocyclic ring or a saccharide residue; the amino acid residue, substituted amino acid residue, nitrogen-containing 5 or 6 membered heterocyclic ring or saccharide residue being optionally joined to the phenyl ring by a linker of an oxygen atom and 1-10 carbon atoms; and an effective amount of at least one metabolic modulator; on a daily basis until an effective level is achieved in blood and/or tumor.
42. The method of claim 41 wherein the compound of Formula I is M4N or maltose-MsN and the metabolic modulator is at least one of Ly294002, etoposide, Rottlerin, dichloroacetate, cetuximab, trastuzumab, bevacizumab and raparaycin.
43, A method for establishing a treatment dosage range or ratio for administration of a combination of a compound of formula 1:
Figure imgf000056_0001
wherein Rj, Rz, R 3 and R4 independently represent hydroxy, a straight or branched chairs lower alkyl or alkoxy, an amino acid residue, a substituted amino acid residue, a nitrogen- containing 5- or 6-membered heterocyclic ring or a saccharide residue; the amino acid residue, substituted amino acid residue, nitrogen-containing 5- or 6-membered heterocyclic ring or saccharide residue being optionally joined to the phenyl ring by a linker of an oxygen atom and 1-10 carbon atoms; and a metabolic modulator; said method comprising conducting titration studies in culture using a TUNEL apoptosis assay and selecting the concentration, concentration range, or ratio that results in adequate apoptosis as the treatment dosage or ratio.
44. The method of claim 41 wherein the compound of Formula I is M4N or maltose-JVhN and the metabolic modulator is at least one of Ly294002, etoposide, Rottlerin, dichloroacetate, cetuximab, trastuzumab, bevacizumab and rapamycin.
PCT/US2009/030459 2008-01-08 2009-01-08 Suppression of cancer growth and metastasis using nordihydroguaiaretic acid derivatives with metabolic modulators WO2009089366A2 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
US15/725,624 USRE46907E1 (en) 2008-01-08 2009-01-08 Suppression of cancer growth and metastasis using nordihydroguaiaretic acid derivatives with metabolic modulators
US12/812,196 US9149526B2 (en) 2008-01-08 2009-01-08 Suppression of cancer growth and metastasis using nordihydroguaiaretic acid derivatives with metabolic modulators
EP09701300.7A EP2240168B1 (en) 2008-01-08 2009-01-08 Suppression of cancer growth and metastasis using nordihydroguaiaretic acid derivatives with metabolic modulators

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US1037108P 2008-01-08 2008-01-08
US61/010,371 2008-01-08
US19182708P 2008-09-12 2008-09-12
US61/191,827 2008-09-12

Publications (2)

Publication Number Publication Date
WO2009089366A2 true WO2009089366A2 (en) 2009-07-16
WO2009089366A3 WO2009089366A3 (en) 2009-10-15

Family

ID=40853765

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2009/030459 WO2009089366A2 (en) 2008-01-08 2009-01-08 Suppression of cancer growth and metastasis using nordihydroguaiaretic acid derivatives with metabolic modulators

Country Status (3)

Country Link
US (2) US9149526B2 (en)
EP (1) EP2240168B1 (en)
WO (1) WO2009089366A2 (en)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011103586A3 (en) * 2010-02-22 2011-12-29 The Johns Hopkins University Suppression of cancer growth and metastasis using nordihydroguaiaretic acid derivatives with 7-hydroxystaurosporine
WO2012032143A1 (en) * 2010-09-10 2012-03-15 Novartis Forschungsstiftung, Zweigniederlassung, Friedrich Miescher Institute For Biomedical Research Phosphorylated twist1 and metastasis
US20150018302A1 (en) * 2012-02-03 2015-01-15 The Johns Hopkins University Compositions comprising ndga derivatives and sorafenib and their use in treatment of cancer
EP2709612A4 (en) * 2011-05-19 2015-07-01 Glax L L C Compositions and methods for treating and preventing cancer by targeting and inhibiting cancer stem cells
CN108342355A (en) * 2018-01-10 2018-07-31 南京艾维艾康生物技术有限公司 Primordial follicle activator and its application in people's ovary cortex culture solution

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
MX2017002227A (en) 2017-02-17 2018-08-16 Promotora Tecnica Ind S A De C V Composition based on nordihydroguaiaretic acid.
EP4065108A4 (en) * 2019-11-25 2023-12-20 The Johns Hopkins University Formulations of terameprocol and temozolomide and their use in stimulation of humoral immunity in tumors

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1995000129A1 (en) 1993-06-23 1995-01-05 Chemex Pharmaceuticals Inc. Treatment of multidrug resistant diseases
WO2003090681A2 (en) 2002-04-24 2003-11-06 Research Development Foundation SYNERGISTIC EFFECTS OF NUCLEAR TRANSCRIPTION FACTOR NF-κB INHIBITORS AND ANTI-NEOPLASTIC AGENTS
WO2003103583A2 (en) 2002-06-10 2003-12-18 Oklahoma Medical Research Foundation A method for using tethered bis(polyhydroxyphenyls) and o-alkyl derivatives thereof in treating inflammatory conditions of the central nervous system
US20070099847A1 (en) 2005-10-28 2007-05-03 The Regents Of The University Of California Tyrosine kinase receptor antagonists and methods of treatment for pancreatic and breast cancer

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4530844A (en) * 1984-07-26 1985-07-23 Warner-Lambert Company Synergistic non-steroidal anti-inflammatory compounds and compositions thereof
MX2007004025A (en) 2004-10-06 2007-06-11 Univ Johns Hopkins Use of nordihydroguaiaretic acid derivatives in the treatment of drug resistant cancer, viral and microbial infection.
EP2538935B1 (en) * 2010-02-22 2014-11-05 The Johns Hopkins University Suppression of cancer growth and metastasis using nordihydroguaiaretic acid derivatives with 7-hydroxystaurosporine

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1995000129A1 (en) 1993-06-23 1995-01-05 Chemex Pharmaceuticals Inc. Treatment of multidrug resistant diseases
WO2003090681A2 (en) 2002-04-24 2003-11-06 Research Development Foundation SYNERGISTIC EFFECTS OF NUCLEAR TRANSCRIPTION FACTOR NF-κB INHIBITORS AND ANTI-NEOPLASTIC AGENTS
WO2003103583A2 (en) 2002-06-10 2003-12-18 Oklahoma Medical Research Foundation A method for using tethered bis(polyhydroxyphenyls) and o-alkyl derivatives thereof in treating inflammatory conditions of the central nervous system
US20070099847A1 (en) 2005-10-28 2007-05-03 The Regents Of The University Of California Tyrosine kinase receptor antagonists and methods of treatment for pancreatic and breast cancer

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
ALTUNDAG K ET AL.: "Overcoming trastuzumab resistance with nordihydroguaiaretic acid?", BREAST CANCER RESEARCH AND TREATMENT, vol. 96, no. 3, 2006, pages 301
BRENNAN P ET AL.: "Inhibition of Nuclear Factor kB by Direct Modification in Whole Cells- Mechanism of Action of Nordihydroguaiaretic Acid, Curcurmin and Thiol Modifiers", BIOCHEMICAL PHARMACOLGY, vol. 55, no. 7, 1998, pages 965 - 973
DANCEY ET AL.: "Strategies for optimising combinations of molecular targeted anticancer agents", NATURE REVIEWS DRUG DISCOVERY, vol. 5, 31 August 2006 (2006-08-31), pages 649 - 659
See also references of EP2240168A4

Cited By (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011103586A3 (en) * 2010-02-22 2011-12-29 The Johns Hopkins University Suppression of cancer growth and metastasis using nordihydroguaiaretic acid derivatives with 7-hydroxystaurosporine
US9101567B2 (en) 2010-02-22 2015-08-11 The Johns Hopkins University Suppression of cancer growth and metastasis using nordihydroguaiaretic acid derivatives with 7-hydroxystaurosporine
WO2012032143A1 (en) * 2010-09-10 2012-03-15 Novartis Forschungsstiftung, Zweigniederlassung, Friedrich Miescher Institute For Biomedical Research Phosphorylated twist1 and metastasis
EP2709612A4 (en) * 2011-05-19 2015-07-01 Glax L L C Compositions and methods for treating and preventing cancer by targeting and inhibiting cancer stem cells
US20150018302A1 (en) * 2012-02-03 2015-01-15 The Johns Hopkins University Compositions comprising ndga derivatives and sorafenib and their use in treatment of cancer
JP2015505567A (en) * 2012-02-03 2015-02-23 ザ・ジョンズ・ホプキンス・ユニバーシティ Compositions comprising NDGA derivatives and sorafenib and their use in the treatment of cancer
CN104640538A (en) * 2012-02-03 2015-05-20 约翰霍普金斯大学 Compositions comprising NDGA derivatives and sorafenib and their use in treatment of cancer
EP2809308A4 (en) * 2012-02-03 2015-08-19 Univ Johns Hopkins Compositions comprising ndga derivatives and sorafenib and their use in treatment of cancer
CN104640538B (en) * 2012-02-03 2018-01-02 约翰霍普金斯大学 Composition comprising NDGA derivatives and Sorafenib and its purposes in treating cancer
US9877978B2 (en) * 2012-02-03 2018-01-30 The Johns Hopkins University Compositions comprising NDGA derivatives and sorafenib and their use in treatment of cancer
CN108342355A (en) * 2018-01-10 2018-07-31 南京艾维艾康生物技术有限公司 Primordial follicle activator and its application in people's ovary cortex culture solution
CN108342355B (en) * 2018-01-10 2021-06-18 南京艾维艾康生物技术有限公司 Primordial follicle activating agent and application thereof in human ovarian cortex culture solution

Also Published As

Publication number Publication date
US9149526B2 (en) 2015-10-06
EP2240168A4 (en) 2011-06-29
USRE46907E1 (en) 2018-06-26
US20110014192A1 (en) 2011-01-20
EP2240168A2 (en) 2010-10-20
EP2240168B1 (en) 2014-05-14
WO2009089366A3 (en) 2009-10-15

Similar Documents

Publication Publication Date Title
EP2240168B1 (en) Suppression of cancer growth and metastasis using nordihydroguaiaretic acid derivatives with metabolic modulators
McKelvey et al. Glycolysis and fatty acid oxidation inhibition improves survival in glioblastoma
Banerjee et al. Retracted: Restoring sensitivity to oxaliplatin by a novel approach in gemcitabine‐resistant pancreatic cancer cells in vitro and in vivo
Zhang et al. RasGAP-derived peptide 38GAP potentiates the cytotoxicity of cisplatin through inhibitions of Akt, ERK and NF-κB in colon carcinoma HCT116 cells
TW201919618A (en) Compounds for treating sarcoma
WO2019109074A1 (en) Mebendazole cancer therapies and methods of use
JP5440985B2 (en) Melanoma treatment
ES2356986T3 (en) ANTITUMOR AGENTS.
AU2015317877A1 (en) Methods and compositions for treating psychotic disorders
Akiyama et al. Edaravone prevents retinal degeneration in adult mice following optic nerve injury
Li et al. Single-Atom Nanocatalytic Therapy for Suppression of Neuroinflammation by Inducing Autophagy of Abnormal Mitochondria
EP2538935B1 (en) Suppression of cancer growth and metastasis using nordihydroguaiaretic acid derivatives with 7-hydroxystaurosporine
WO2018237174A2 (en) Cystic fibrosis transmembrane conductance regulator modulators for treating autosomal dominant polycystic kidney disease
US20100144652A1 (en) Composition comprising a glycolytic inhibitor and a ring system comprising a sulphamate group for the treatment of cancer
RU2472512C1 (en) Antituberculous composition and method for preparing it
JP6205373B2 (en) Compositions comprising NDGA derivatives and sorafenib and their use in the treatment of cancer
CN113677202B (en) Mitochondrial targeting isoparaffin/ISOLEVUGLANDIN scavengers
US11065230B2 (en) Methods and pharmaceutical compositions for the treatment of systemic mastocytosis
WO2018136933A1 (en) Inhibition of stromal interaction molecule 1 (stim1) as a co-treatment for adult onset polycystic kidney disease (adpkd)
Yu 2-Methoxy-6-Acetyl-7-Methyljuglone (MAM), a Natural Product, Kills Cancer Cells by Targeting NQO1
WO2022253748A1 (en) Combinations for the treatment of hpv
WO2023230212A1 (en) Combination treatment for adenocarcinoma
Fan et al. Activated SIRT1 and SIRT6 by Nrf2 Activator Attenuated Cisplatin-Induced Acute Nephrotoxicity in Mice
WO2023159048A2 (en) Small molecule stat3 inhibitor for treating triple negative breast cancer
WO2024103012A1 (en) Polycyclic compounds for use in neurodegenerative conditions

Legal Events

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

Ref document number: 09701300

Country of ref document: EP

Kind code of ref document: A2

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2009701300

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 12812196

Country of ref document: US