WO2008150506A1 - Pulsatile dosing of gossypol for treatment of disease - Google Patents

Pulsatile dosing of gossypol for treatment of disease Download PDF

Info

Publication number
WO2008150506A1
WO2008150506A1 PCT/US2008/006936 US2008006936W WO2008150506A1 WO 2008150506 A1 WO2008150506 A1 WO 2008150506A1 US 2008006936 W US2008006936 W US 2008006936W WO 2008150506 A1 WO2008150506 A1 WO 2008150506A1
Authority
WO
WIPO (PCT)
Prior art keywords
gossypol
administered
carcinoma
cancer
apoptosis
Prior art date
Application number
PCT/US2008/006936
Other languages
French (fr)
Inventor
Jon T. Holmlund
Mel Sorensen
Lance Leopold
Dajun Yang
Original Assignee
Ascenta Therapeutics, Inc.
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 Ascenta Therapeutics, Inc. filed Critical Ascenta Therapeutics, Inc.
Priority to AU2008260510A priority Critical patent/AU2008260510A1/en
Priority to EP08768032A priority patent/EP2152076A4/en
Priority to CA002689033A priority patent/CA2689033A1/en
Priority to JP2010510372A priority patent/JP2010529023A/en
Publication of WO2008150506A1 publication Critical patent/WO2008150506A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/11Aldehydes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/337Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having four-membered rings, e.g. taxol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • 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
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/18Drugs for disorders of the alimentary tract or the digestive system for pancreatic disorders, e.g. pancreatic enzymes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/08Drugs for disorders of the urinary system of the prostate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/10Drugs for disorders of the urinary system of the bladder
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P33/00Antiparasitic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • This invention is in the field of medicinal chemistry.
  • the invention relates to pulsatile dose administration of gossypol or pharmaceutical compositions thereof for treating diseases, disorders and conditions responsive to gossypol, inhibiting the activity of anti-apoptotic Bcl-2 family proteins, inducing apoptosis in cells and increasing the sensitivity of cells to inducers of apoptosis.
  • the aggressive cancer cell phenotype is the result of a variety of genetic and epigenetic alterations leading to deregulation of intracellular signaling pathways (Ponder, Nature 411:336 (2001)).
  • the commonality for all cancer cells is their failure to execute an apoptotic program, and lack of appropriate apoptosis due to defects in the normal apoptosis machinery is a hallmark of cancer (Lowe et ai, Carcinogenesis 27:485 (2000)).
  • Most of the current cancer therapies including chemotherapeutic agents, radiation (radiotherapeutic agents), and immunotherapy, work by indirectly inducing apoptosis in cancer cells.
  • IAPs apoptosis proteins
  • the second class of central negative regulators of apoptosis is the BcI-
  • Bcl-2 is the founding member of the family and was first isolated as the product of an oncogene.
  • the Bcl-2 family now includes both anti-apoptotic molecules such as Bcl-2 and BCI-X L and pro-apoptotic molecules such as Bax, Bak, Bid, and Bad.
  • Bcl-2 and BCI-X L are overexpressed in many types of human cancer ⁇ e.g., breast, prostate, colorectal, lung, etc.), including Non-Hodgkin's lymphoma, which is caused by a chromosomal translocation (tl4, 18) that leads to overexpression of Bcl-2.
  • tl4, 18 chromosomal translocation
  • Bcl-2 and BCI-X L are thought to play a role in tumor cell migration and invasion, and therefore, metastasis (Amberger et al., Cancer Res. 55:149 (1998); Wick et al, FEBS Lett. 440:419 (1998); Mohanam et al., Cancer Res. 53:4143 (1993); Pedersen et al, Cancer Res. 55:5158 (1993)).
  • Bcl-2 family proteins appear to provide tumor cells with a mechanism for surviving in new and non-permissive environments (e.g., metastatic sites), and contribute to the organospecific pattern of clinical metastatic cancer spread (Rubio, Lab Invest. 81:725 (2001); Fernandez et al, Cell Death Differ.
  • Anti- apoptotic proteins such as Bcl-2 and/or BCI-X L are also thought to regulate cell-cell interactions, for example through regulation of cell surface integrins (Reed, Nature 387:773 (1997); Frisch et al, Curr. Opin. Cell Biol. 9:701 (1997); Del Bufalo et al. , FASEB J. 11:947 (1997)).
  • Gossypol is a naturally occurring double biphenolic compound derived from crude cotton seed oil (Gossypium sp.). Human trials of gossypol as a male contraceptive have demonstrated the safety of long term administration of these compounds (Wu, Drugs 38:333 (1989)). Gossypol has more recently been shown to have some antiproliferative effects (Flack et al., J. Clin. Endocrinol. Metab. 76:1019 (1993); Bushunow et al., J. Neuro-Oncol. 43:79, (1999); Van Poznak et al, Breast Cancer Res. Treat. 66:239 (2001)). Gossypol and its derivatives recently have been shown to be potent inhibitors of Bcl-2 and BCI-X L and to have strong anti-cancer activity (U.S. Patent Application No. 2003/0008924).
  • the present invention relates to pulsatile dose administration of gossypol, i.e., ( ⁇ )-gossypol (I), (-)-gossypol (II) or (+)-gossypol. It has surprisingly been found that pulsatile dose administration of gossypol provides clinical efficacy coupled with a reduction in adverse events. Gossypol and pharmaceutical compositions thereof are useful for treating hyperproliferative and other diseases, inhibiting the activity of anti-apoptotic Bcl-2 family proteins, inducing apoptosis in cells and increasing the sensitivity of cells to inducers of apoptosis.
  • the present invention contemplates that pulsatile dose administration of gossypol to patients suffering from cancer and other diseases will expose patients to therapeutically effective amounts of gossypol and will minimize unwanted adverse events.
  • Gossypol inhibits the function(s) of anti-apoptotic Bcl-2 family proteins and will kill cancer cells or supporting cells outright (those cells whose continued survival is dependent on the overactivity of Bcl-2 family proteins) and/or render such cells as a population more susceptible to the cell death-inducing activity of chemotherapeutic and/or radiotherapeutic agents.
  • the present invention contemplates that gossypol administered by pulsatile dosing will satisfy an unmet need for the treatment of multiple cancer types, either when administered as monotherapy to induce apoptosis in cancer cells dependent on anti-apoptotic Bcl-2 family proteins function, or when administered in a temporal relationship with other cell death-inducing chemotherapeutic and/or radiotherapeutic agents so as to render a greater proportion of the cancer cells or supportive cells susceptible to executing the apoptosis program compared to the corresponding proportion of cells in a patient treated only with the chemotherapeutic and/or radiotherapeutic agent alone.
  • the present invention further contemplates that gossypol administered by pulsatile dosing will induce apoptosis and/or render cells more sensitive to induction of apoptosis in other diseases or conditions characterized by dysregulation of apoptosis.
  • a therapeutically effective amount of gossypol administered by pulsatile dosing and one or more additional therapeutic agents will produce a greater tumor response and clinical benefit in such patients compared to those treated with either gossypol or one or more therapeutic agents alone.
  • gossypol administered by pulsatile dosing lowers the apoptotic threshold of all cells that express anti- apoptotic Bcl-2 family proteins, the proportion of cells that successfully execute the apoptosis program in response to the apoptosis inducing activity of therapeutic agents, such as anticancer drugs, will be increased.
  • gossypol administered by pulsatile dosing is expected to allow administration of a lower, and therefore less toxic and more tolerable, dose of an anticancer agent to produce the same tumor response/clinical benefit as the conventional dose of the anticancer agent alone. Since the doses for all approved anticancer agents are known, the present invention contemplates combination therapies with various combinations of known anticancer agents with gossypol administered by pulsatile dosing.
  • gossypol administered by pulsatile dosing acts at least in part by inhibiting anti-apoptotic Bcl-2 family proteins
  • the exposure of cancer cells and supporting cells to therapeutically effective amounts of gossypol can be temporally linked to coincide with the attempts of cells to execute the apoptosis program in response to the anticancer agent.
  • administering gossypol by pulsatile dosing in connection with certain temporal relationships will provide especially efficacious therapeutic practices.
  • Gossypol administered by pulsatile dosing is useful for the treatment, amelioration, or prevention of disorders responsive to induction of apoptotic cell death, e.g., disorders characterized by dysregulation of apoptosis, including hyperproliferative diseases such as cancer.
  • gossypol administered by pulsatile dosing can be used to treat, ameliorate, or prevent cancer that is characterized by resistance to cancer therapies (e.g., those which are chemoresistant, radiation resistant, hormone resistant, and the like).
  • the cancer is chemoresistant to treatment with a taxane, i.e., docetaxel or paclitaxel.
  • the chemoresistant cancer is prostate cancer.
  • gossypol administered by pulsatile dosing can be used to treat, ameliorate, or prevent metastatic cancer.
  • gossypol administered by pulsatile dosing can be used to treat hyperproliferative and other diseases characterized by overexpression of anti-apoptotic Bcl-2 family proteins.
  • gossypol administered by pulsatile doing can be used to modulate spermicidal activity (e.g., function as a male contraceptive or antifertility agent), treat malaria, microbial or viral disease (e.g., inhibit the growth of the HIV virus as a treatment for AIDS), treat obesity, skin disorders or baldness, inhibit growth of endothelial cells, inhibit vascularization or neovascularization, treat arthritic conditions, neovascular-based dermatological conditions, diabetic retinopathy, Kaposi's sarcoma, age-related macular degeneration, restenosis, telangiectasia, glaucoma, keloids, corneal graft rejection, wound granularization, angiofibroma, Osier-Webber syndrome, myocardial angiogenesis, psoriatic arthritis or scleroderma, inhibit DNA synthesis or DNA polymerase activity and treat gynecological disorders (e.g., endo
  • Fig. 1 is a chart showing pulsatile dose administration schedules of gossypol.
  • Fig. 2 is a graph showing the in vivo activity of (-)-gossypol acetic acid co-crystal via pulse dose administration in combination with docetaxel.
  • Fig. 3 is a graph showing the in vivo activity of (-)-gossypol acetic acid co-crystals via pulse dose administration in combination with docetaxel.
  • Fig, 4 is a waterfall plot showing PSA response in chemo-naive patients after treatment with (-)-gossypol acetic acid co-crystals.
  • Fig. 5 is a graph showing PSA response versus time in chemo-naive patients after treatment with (-)-gossypol acetic acid co-crystals.
  • Fig. 6 is a series of four images showing tumor size before and after treatment with (-)-gossypol acetic acid co-crystals.
  • Fig. 7 is a waterfall plot showing PSA response in docetaxel (taxotere)- resistant patients after treatment with (-)-gossypol acetic acid co-crystals.
  • Fig. 8 is a graph showing PSA response kinetics in a docetaxel
  • Fig. 9 is a graph showing PSA response kinetics in a docetaxel
  • Fig. 10 is a graph showing PSA response kinetics in a docetaxel
  • the present invention relates to pulsatile dose administration of gossypol.
  • Various compositions comprising gossypol are contemplated for use in the methods of the invention.
  • a gossypol composition may comprise, for example, ( ⁇ )-gossypol, (-)-gossypol, (+)-gossypol, ( ⁇ )-gossypol co- crystals, (-)-gossypol co-crystals or (+)-gossypol co-crystals.
  • Gossypol is an inhibitor of anti-apoptotic Bcl-2 family proteins.
  • the invention relates to methods of sensitizing cells to inducers of apoptosis and to methods of inducing apoptosis in cells, comprising administering gossypol by pulsatile dosing alone or in combination with an additional therapeutic agent, such as an inducer of apoptosis.
  • the invention further relates to methods of treating, ameliorating, or preventing disorders in a patient that are responsive to induction of apoptosis comprising administering to the patient gossypol by pulsatile dosing and an inducer of apoptosis, e.g., an anticancer agent,
  • disorders include those characterized by a dysregulation of apoptosis and those characterized by overexpression of anti-apoptotic Bcl-2 family proteins.
  • the disease, condition or disorder responsive to the induction of apoptosis is selected from the group consisting of a hyperproliferative disease, i.e., cancer autoimmune disorder, chronic inflammatory condition, i.e., psoriasis, viral infection, microbial infection, parasitic infection, osteoarthritis, and atherosclerosis.
  • the cancer is selected from the group consisting of breast cancer, prostate cancer, lymphoma, skin cancer, pancreatic cancer, colon cancer, malignant melanoma, ovarian cancer, brain cancer, primary brain carcinoma, head-neck cancer, glioma, glioblastoma, liver cancer, bladder cancer, non-small cell lung cancer, head carcinoma, neck carcinoma, breast carcinoma, ovarian carcinoma, lung carcinoma, small-cell lung carcinoma, Wilms' tumor, cervical carcinoma, testicular carcinoma, bladder carcinoma, pancreatic carcinoma, stomach carcinoma, colon carcinoma, prostatic carcinoma, genitourinary carcinoma, thyroid carcinoma, esophageal carcinoma, myeloma, multiple myeloma, adrenal carcinoma, renal cell carcinoma, endometrial carcinoma, adrenal cortex carcinoma, malignant pancreatic insulinoma, malignant carcinoid carcinoma, choriocarcinoma, mycosis fungoides, malignant hypercalcemia, cervical hyperplasia, leukemia, acute lymphocytic leukemia,
  • gossypol refers to ( ⁇ )-gossypol
  • (-)-gossypol refers to an optically active composition of gossypol wherein the active molecules comprising the composition rotate plane polarized light counterclockwise (e.g., levorotatory molecules) as measured by a polarimeter.
  • the (-)-gossypol compound has an enantiomeric excess of 1% to 100%.
  • the (-)-gossypol compound has an enantiomeric excess of at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% (-)-gossypol.
  • the specific rotation ([ ⁇ ]o) of the compound is about -350° to about -390°, about -375° to about -390°, or about -385° to about -390°.
  • C 1-8 carboxylic acid refers to straight- chained or branched, aromatic or non-aromatic, saturated or unsaturated, substituted or unsubstituted C 1-8 carboxylic acid, including, but not limited to, formic acid, acetic acid, propionic acid, n-butyric acid, t-butyric acid, n- pentanoic acid, 2-pentanoic acid, n-hexanoic acid, 2-hexanoic acid, n- heptanoic acid, n-octanoic acid, acrylic acid, succinic acid, fumaric acid, malic acid, tartaric acid, citric acid, lactic acid, and benzoic acid.
  • Ci -8 sulfonic acid refers to straight-chained or branched, aromatic or non-aromatic, saturated or unsaturated, substituted or unsubstituted C 1-8 sulfonic acid, including, but not limited to, methanesulfonic acid, ethanesulfonic acid, n-propanesulfonic acid, 2-propanesulfonic acid, n- butanesulfonic acid, n-pentanesulfonic acid n-hexanesulfonic acid, n- heptanesulfonic acid, n-octanesulfonic acid, and benzenesulfonic acid.
  • C 1-I2 ketone refers to straight-chained, cyclic or branched, aromatic or non-aromatic, saturated or unsaturated, substituted or unsubstituted Ci -12 ketone, including, but not limited to, acetone and cyclododecanone.
  • Ci -12 diketone refers to straight-chained or branched, aromatic or non-aromatic, saturated or unsaturated, substituted or unsubstituted C 1-12 diketone, including, but not limited to, 2,4-pentanedione.
  • the term "gossypol co-crystal,” as used herein, refers to a composition comprising co-crystals of ( ⁇ )-gossypol and a Ci -8 carboxylic acid, C 1-8 sulfonic acid, C M2 ket one or C M2 diketone.
  • the term "(-)-gossypol co- crystal,” as used herein, refers to a composition comprising (-)-gossypol and acetic acid, (-)-gossypol and acetone, (-)-gossypol and 2,4-pentanedione or (-)- gossypol and cyclododecanone.
  • the (-)-gossypol co- crystal is (-)-gossypol acetic acid co-crystal. See U.S. 7,342,046. A method of preparing (-)-gossypol acetic acid co-crystal is described in U.S. 2008/0021110.
  • Pulsatile dose administration refers to intermittent (i.e., not constant daily) administration of gossypol.
  • Pulsatile dose administration schedules useful in the present invention encompass any discontinuous daily administration regimen that provides a therapeutically effective amount of gossypol to a patient in need thereof. Pulsatile dosing regimens may use equivalent, lower or higher doses of gossypol than typically used in continuous daily dosing regimens. On the days that gossypol is scheduled to be administered, administration of gossypol may occur once a day, twice-a-day (i.e., BID), three times a day, four times a day or more in accordance with an intermittent daily dosing schedule.
  • BID twice-a-day
  • the therapeutic utility of drug administration can be offset by the number and severity of adverse events a patient experiences.
  • Pulsatile dosing of gossypol results in the unexpected combination of a reduction in the number and/or severity of clinical adverse events coupled with a maintenance or enhancement in clinical efficacy, as compared to continuous daily dosing.
  • the surprising clinical benefits of pulsatile dose administration of gossypol may be even more prominent when combined with other therapeutic agents.
  • gossypol may be administered no more frequently than one day out of every two days (i.e., day 1, day 3, day 5, day 7, day 9, etc.), every three days (i.e., day 1, day 4, day 7, day 10, etc.), every four days, every five days, every six days, every seven days, every eight days, every nine days, every ten days, every two weeks, every three weeks, every four weeks, or longer.
  • the administration of gossypol can continue for one, two, three or four weeks, one, two, three or four months, one, two, three or four years or longer.
  • gossypol may be administered on a least two consecutive days, e.g., at least three, four, five, six or seven consecutive days, followed by at least one day, at least two consecutive days, at least three consecutive days, at least four consecutive days, at least five consecutive days, at least six consecutive days, at least seven consecutive days, at least eight consecutive days, at least nine consecutive days, at least ten consecutive days, at least eleven consecutive days, at least twelve consecutive days, at least thirteen consecutive days, at least two consecutive weeks, at least three consecutive weeks or at least four consecutive weeks or longer wherein gossypol is not administered.
  • the administration of gossypol can continue for one, two, three or four weeks, one, two, three or four months, one, two, three or four years or longer.
  • gossypol may be administered twice-a-day for three consecutive days followed by eleven, twelve, thirteen, fourteen, fifteen, sixteen, seventeen, eighteen, nineteen or twenty consecutive days, or longer, wherein gossypol is not administered.
  • a pharmaceutical composition comprising (-)-gossypol may be administered twice-a-day for three consecutive days followed by seventeen or eighteen consecutive days wherein gossypol is not administered. The administration of gossypol can continue for two, three or four weeks, one, two, three or four months, one, two, three or four years or longer.
  • gossypol may be administered one day a week, e.g., gossypol administered on one day followed by six consecutive days wherein gossypol is not administered, for at least two weeks, at least three weeks, at least four weeks, at least five weeks, at least six weeks, at least seven weeks or at least eight weeks.
  • the administration of gossypol can continue for one, two, three or four months, one, two, three or four years or longer.
  • gossypol may be administered via the sequential use of a combination of two or more pulsatile dosing schedules.
  • the combination may comprise the same pulsatile dosing schedules, e.g., gossypol administered twice-a-day for three consecutive days followed by eleven consecutive days wherein gossypol is not administered followed by gossypol administered twice-a-day for three consecutive days followed by eleven consecutive days wherein gossypol is not administered, or different pulsatile dosing schedules, e.g., gossypol administered twice-a-day for three consecutive days followed by eleven consecutive days wherein gossypol is not administered followed by gossypol administered on one day followed by six consecutive days wherein gossypol is not administered.
  • pulsatile dose administration of gossypol comprises administration on one day followed by six consecutive days wherein gossypol is not administered followed by administration on at least two consecutive days followed by at least one day wherein gossypol is not administered.
  • pulsatile dose administration of gossypol comprises administration twice-a- day for at least three consecutive days followed by at least seven consecutive days wherein gossypol is not administered followed by administration on one day followed by at least six consecutive days wherein gossypol is not administered.
  • pulsatile dose administration of gossypol comprises administration on one day followed by six consecutive days wherein gossypol is not administered followed by administration on one day followed by one day wherein gossypol is not administered.
  • the sequential use of a combination of two or more pulsatile dosing regimens may be repeated as many times as necessary to achieve or maintain a therapeutic response, e.g., from one to about fifty times, e.g., from one to about twenty times, e.g., from about one to about ten times. With every repetition any additional therapeutic agents may be the same or different from that used in the previous repetition.
  • gossypol may be administered according to a pulsatile dosing schedule and/or sequential combination of two or more pulsatile dosing schedules followed by a waiting period.
  • the term "waiting period,” as used herein, refers to a period of time between dosing schedules when gossypol is not administered to the patient.
  • the waiting period may be one, two, three, four, five or six days, one, two or three weeks, one, two, three or four months, one, two, three or four years or longer.
  • the waiting period may be one to thirty days, e.g., seven, fourteen, twenty one or thirty days, e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29 or 30 days.
  • the same or a different pulsatile dosing schedule and/or sequential combination of one or more pulsatile dosing schedules of gossypol may resume.
  • pulsatile dose administration of gossypol comprises administration on one day followed by six consecutive days wherein gossypol is not administered, a waiting period and administration on at least two consecutive days followed by at least one day wherein gossypol is not administered.
  • pulsatile administration of gossypol comprises administration on one day followed by six consecutive days wherein gossypol is not administered, a waiting period and administration on one day followed by six consecutive days wherein gossypol is not administered.
  • pulsatile dose administration of gossypol comprises administration twice-a-day for at least three consecutive days followed by at least seven consecutive days wherein gossypol is not administered, a waiting period and administration on one day followed by at least six consecutive days wherein gossypol is not administered.
  • the pulsatile dosing/waiting period regimen may be repeated as many times as necessary to achieve or maintain a therapeutic response, e.g., from one to about fifty times, e.g., from one to about twenty times, e.g., from about one to about ten times. With every repetition any additional therapeutic agents may be the same or different from that used in the previous repetition.
  • gossypol may be administered, for example, via the sequential use of a combination of schedule A and B, schedule B and C, schedule C and D, schedule D and E, schedule F and G, schedule G and H, schedule H and I, schedule H and H, schedule I and I, schedule H, I and H or schedule G, H and I.
  • gossypol may be administered via schedule H followed by a waiting period ⁇ e.g., one to thirty days) followed by administration via schedule I, schedule A-waiting period- schedule B, schedule H- waiting period-schedule E- waiting period-schedule A, schedule H-waiting period-schedule I-waiting period-schedule H or schedule H-schedule I-waiting period-schedule H-schedule I.
  • the above-described pulsatile dose administration schedules are provided for illustrative purposes only and should not be considered limiting.
  • the present invention contemplates any discontinuous daily administration regimen that provides a therapeutically effective amount of gossypol to a patient in need thereof.
  • adverse event refers to any undesirable change in health that occurs to a patient during a clinical trial or within a period of time after the clinical trial is complete.
  • Adverse events are categorized by grade, with less serious adverse events given grades 1 (mild) and 2 (moderate) and more serious adverse events given grades 3 (severe) and 4 (life-threatening or disabling).
  • the grading of adverse events can be done using any scale known in the art, such as the National Cancer Institute scale (Common Terminology Criteria for Adverse Events, v3.0).
  • a decrease in the number of adverse events refers to a decrease in the actual number of events.
  • a decrease in the severity of adverse events refers to a decrease in the grade of the adverse events that occur.
  • Bcl-2 family proteins refers to both the anti-apoptotic members of the Bcl-2 family, including, but not limited to, BcI- 2, BcI-XL, McI-I, Al/BFL-1, BOO-DIVA, Bcl-w, Bcl-6, Bcl-8, and Bcl-y, and the pro-apoptotic members of the Bcl-2 family, including, but not limited to, Bak, Bax, Bad, tBid, Hrk, Bim, Bmf, as well as other Bcl-2 homology domain 3 (BH3) containing proteins that are regulated by gossypol compounds.
  • BcI- 2 BcI- 2
  • McI-I McI-I
  • Al/BFL-1 BOO-DIVA
  • Bcl-w Bcl-6
  • Bcl-8 Bcl-y
  • pro-apoptotic members of the Bcl-2 family including, but not limited to, Bak, Bax, Bad, tB
  • anti-apoptotic Bcl-2 family proteins refers to an elevated level (e.g., aberrant level) of mRNAs encoding for an anti-apoptotic Bcl-2 family protein(s), and/or to elevated levels of anti-apoptotic Bcl-2 family protein(s) in cells as compared to similar corresponding non-pathological cells expressing basal levels of mRNAs encoding anti-apoptotic Bcl-2 family proteins or having basal levels of anti- apoptotic Bcl-2 family proteins.
  • Methods for detecting the levels of mRNAs encoding anti-apoptotic Bcl-2 family proteins or levels of anti-apoptotic Bcl-2 family proteins in a cell include, but are not limited to, Western blotting using anti-apoptotic Bcl-2 family protein antibodies, immunohistochemical methods, and methods of nucleic acid amplification or direct RNA detection.
  • Western blotting using anti-apoptotic Bcl-2 family protein antibodies immunohistochemical methods, and methods of nucleic acid amplification or direct RNA detection.
  • As important as the absolute level of anti-apoptotic Bcl-2 family proteins in cells is to determining that they overexpress anti-apoptotic Bcl-2 family proteins, so also is the relative level of anti-apoptotic Bcl-2 family proteins to other pro- apoptotic signaling molecules (e.g., pro-apoptotic Bcl-2 family proteins) within such cells.
  • the pro-apoptotic signaling molecules would be sufficient to cause the cells to execute the apoptosis program and die, said cells would be dependent on the anti-apoptotic Bcl-2 family proteins for their survival.
  • exposure to an inhibiting effective amount of an anti-apoptotic Bcl-2 family protein inhibitor will be sufficient to cause the cells to execute the apoptosis program and die.
  • an anti-apoptotic Bcl-2 family protein also refers to cells that, due to the relative levels of pro-apoptotic signals and anti- apoptotic signals, undergo apoptosis in response to inhibiting effective amounts of compounds that inhibit the function of anti-apoptotic Bcl-2 family proteins.
  • a therapeutic agent refers to any agent which can be used in the treatment, management, or amelioration of a disease, condition or disorder or one or more symptoms thereof.
  • the term “therapeutic agent” refers to gossypol, e.g., (-)-gossypol. In certain other embodiments, the term “therapeutic agent” does not refer to gossypol.
  • a therapeutic agent is an agent which is known to be useful for, or has been or is currently being used for the treatment, management, prevention, or amelioration of a disorder or one or more symptoms thereof.
  • the therapeutic agent is an anticancer agent.
  • anticancer agent and “anticancer drug” as used herein, refer to any therapeutic agent (e.g., chemotherapeutic compounds and/or molecular therapeutic compounds), radiation therapies, or surgical interventions, used in the treatment of hyperproliferative diseases such as cancer (e.g., in mammals).
  • therapeutic agent e.g., chemotherapeutic compounds and/or molecular therapeutic compounds
  • radiation therapies e.g., radiation therapies
  • surgical interventions used in the treatment of hyperproliferative diseases such as cancer (e.g., in mammals).
  • a therapeutically effective amount refers to that amount of the therapeutic agent sufficient to result in amelioration of one or more symptoms of a disorder, or prevent advancement of a disorder, or cause regression of the disorder.
  • a therapeutically effective amount preferably refers to the amount of a therapeutic agent that decreases the rate of tumor growth, decreases tumor mass, decreases the number of metastases, increases time to tumor progression, or increases survival time by at least 5%, preferably at least 10%, at least 15%, at least 20%, at least 25%, at least 30%, at least 35%, at least 40%, at least 45%, at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, or at least 100%.
  • sensitize and “sensitizing,” as used herein, refer to making, through the administration of gossypol, a patient or a cell within a patient more susceptible, or more responsive, to the biological effects (e.g., promotion or retardation of an aspect of cellular function including, but not limited to, cell growth, proliferation, invasion, angiogenesis, or apoptosis) of a second therapeutic agent.
  • biological effects e.g., promotion or retardation of an aspect of cellular function including, but not limited to, cell growth, proliferation, invasion, angiogenesis, or apoptosis
  • the sensitizing effect of gossypol on a target cell can be measured as the difference in the intended biological effect (e.g., promotion or retardation of an aspect of cellular function including, but not limited to, cell growth, proliferation, invasion, angiogenesis, or apoptosis) observed upon the administration of a second therapeutic agent with and without administration of gossypol.
  • the intended biological effect e.g., promotion or retardation of an aspect of cellular function including, but not limited to, cell growth, proliferation, invasion, angiogenesis, or apoptosis
  • the response of the sensitized cell can be increased by at least 5 %, at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 100%, at least 150%, at least 200%, at least 350%, at least 300%, at least 350%, at least 400%, at least 450%, or at least 500% over the response in the absence of gossypol.
  • the term "dysregulation of apoptosis,” as used herein, refers to any aberration in the ability of (e.g., predisposition) a cell to undergo cell death via apoptosis.
  • Dysregulation of apoptosis is associated with or induced by a variety of conditions, including for example, autoimmune disorders (e.g., systemic lupus erythematosus, rheumatoid arthritis, graft-versus-host disease, myasthenia gravis, or Sjogren's syndrome), chronic inflammatory conditions (e.g., psoriasis, asthma or Crohn's disease), hyperproliferative disorders (e.g., tumors, B cell lymphomas, or T cell lymphomas), viral infections (e.g., herpes, papilloma, or HIV), microbial infections, parasitic infections and other conditions such as osteoarthritis and atherosclerosis.
  • the dysregulation is induced by or associated with a viral infection, the viral infection may or may not be detectable at the time dysregulation occurs or is observed. That is, viral-induced dysregulation can occur even after the disappearance of symptoms of viral infection.
  • hyperproliferative disease refers to any condition in which a localized population of proliferating cells in an animal is not governed by the usual limitations of normal growth, such as cancer.
  • hyperproliferative disorders include psoriasis, restenosis, tumors, neoplasms, lymphomas and the like.
  • a neoplasm is said to be benign if it does not undergo invasion or metastasis and malignant if it does either of these.
  • a "metastatic" cell means that the cell can invade and destroy neighboring body structures.
  • Hyperplasia is a form of cell proliferation involving an increase in cell number in a tissue or organ without significant alteration in structure or function.
  • Metaplasia is a form of controlled cell growth in which one type of fully differentiated cell substitutes for another type of differentiated cell.
  • cancer is intended to refer to any known cancer, and may include, but is not limited to the following: leukemias such as acute leukemia, acute lymphocytic leukemia, acute myelocytic leukemias such as myeloblastic, promyelocytic, myelomonocytic, monocytic, and erythroleukemia leukemias, and myelodysplastic syndrome; chronic leukemias such as chronic myelocytic (granulocytic) leukemia, chronic lymphocytic leukemia, and hairy cell leukemia; polycythemia vera; lymphomas such as Hodgkin's disease and non-Hodgkin's disease; multiple myelomas such as smoldering multiple myeloma, non-secretory myeloma, osteosclerotic myeloma, plasma cell leukemia, solitary plasmacytoma and extramedullary
  • myxosarcoma osteogenic sarcoma
  • endotheliosarcoma endotheliosarcoma
  • lymphangioendotheliosarcoma mesothelioma
  • synovioma hemangioblastoma
  • epithelial carcinoma cystadenocarcinoma
  • cystadenocarcinoma bronchogenic carcinoma
  • sweat gland carcinoma sebaceous gland carcinoma
  • autoimmune disorder refers to any condition in which an organism produces antibodies or immune cells which recognize the organism's own molecules, cells or tissues.
  • Non-limiting examples of autoimmune disorders include autoimmune hemolytic anemia, autoimmune hepatitis, Berger's disease or IgA nephropathy, celiac sprue, chronic fatigue syndrome, Crohn's disease, dermatomyositis, fibromyalgia, graft versus host disease, Grave's disease, Hashimoto's thyroiditis, idiopathic thrombocytopenia purpura, lichen planus, multiple sclerosis, myasthenia gravis, psoriasis, rheumatic fever, rheumatic arthritis, scleroderma, Sjogren's syndrome, systemic lupus erythematosus, type 1 diabetes, ulcerative colitis, vitiligo, and the like.
  • neoplastic disease refers to any abnormal growth of cells being either benign (non-cancerous) or malignant (cancerous).
  • anti-neoplastic agent refers to any compound that retards the proliferation, growth, or spread of a targeted ⁇ e.g., malignant) neoplasm.
  • prevention refers to a decrease in the occurrence of pathological cells ⁇ e.g., hyperproliferative or neoplastic cells) in an animal.
  • the prevention may be complete, e.g., the total absence of pathological cells in a subject.
  • the prevention may also be partial, such that the occurrence of pathological cells in a subject is less than that which would have occurred without the present invention.
  • the term "synergistic,” as used herein, refers to an effect obtained when gossypol and a second therapeutic agent are administered together ⁇ e.g., at the same time or one after the other) that is greater than the additive effect of gossypol and the second therapeutic agent when administered individually.
  • the synergistic effect allows for lower doses of gossypol and/or the second therapeutic agent to be administered or provides greater efficacy at the same doses.
  • the synergistic effect obtained can be at least 5 %, at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 100%, at least 125%, at least 150%, at least 175%, at least 200%, at least 250%, at least 300%, at least 350%, at least 400%, or at least 500% more than the additive effect of gossypol and the second therapeutic agent when administered individually.
  • the synergistic effect can be a decrease in the rate of tumor growth, a decrease in tumor mass, a decrease in the number of metastases, an increase in time to tumor progression, or an increase in survival time.
  • the co-administration of gossypol by pulsatile dosing and an anticancer agent may allow for the use of lower doses of gossypol and/or the anticancer agent such that the cancer is effectively treated while avoiding any substantial toxicity to the subject.
  • Inhibitors of anti-apoptotic Bcl-2 family proteins useful in the present invention include, for example, gossypol or co-crystals of gossypol or pharmaceutical compositions thereof that are administered by pulsatile dosing.
  • gossypol or co-crystals of gossypol or pharmaceutical compositions thereof that are administered by pulsatile dosing.
  • Compositions comprising co-crystals of gossypol may be more stable than some other compositions comprising gossypol.
  • Any Ci -8 carboxylic acid or Cj -8 sulfonic acid that is capable of stabilizing gossypol may be used in the composition.
  • compositions comprising gossypol useful in this invention may be prepared using methods known to those of skill in the art, such as the methods disclosed in U.S. Published Application No. 2005/0234135.
  • (-)- gossypol co-crystal may be prepared by dissolving (-)-gossypol in acetone to form a solution, filtering the solution, adding acetic acid into the solution with mixing until the solution turns turbid, leaving the turbid solution at room temperature and then at reduced temperature to form co-crystals, collecting the co-crystals, washing the co-crystals with a solvent, and drying the co-crystals.
  • Reduced temperature is less than about 2O 0 C, preferably about 0-15 0 C, more preferably about 4°C.
  • the time for co-crystal formation may range from 1 hour to 1 day; preferably the time is about 1-4 hours.
  • the co-crystals may be collected by any suitable means, including by filtration.
  • the solvent for washing the co-crystals may be any suitable solvent, e.g., hexane, pentane, benzene, toluene, or petroleum ether.
  • the washed co-crystals may be dried at room temperature, preferably in a lightproof container.
  • the co-crystals may also be dried in a vacuum drier, preferably at an elevated temperature ⁇ e.g., about 30-60 0 C, more preferably about 4O 0 C) for about 6-72 hours, preferably about 12-48 hours.
  • Gossypol acetic acid co-crystals may also be prepared from a mixture of ( ⁇ )-gossypol and acetic acid via recrystallization.
  • the gossypol acetic acid co-crystals may be further recrystallized from a solution of gossypol acetic acid in a mixture of acetone and acetic acid.
  • the recrystallization mixture is held for about 15 minutes to about 100 minutes, e.g., about 30 minutes to about 60 minutes, to allow co-crystal formation.
  • the recrystallization is carried out at ambient temperature, e.g., about 15 0 C to about 3O 0 C, e.g., about 22 0 C.
  • the crystals are harvested from the recrystallization mixture (e.g., by filtration) and washed with a non-polar solvent, e.g., pentane, hexene, hexane(s), heptane, or mixtures thereof.
  • a non-polar solvent e.g., pentane, hexene, hexane(s), heptane, or mixtures thereof.
  • the washing step is quick to avoid the incorporation of the non- polar solvent into the crystals. Short washing times (less than 2 minutes) are preferred.
  • the crystals may then be dried, e.g., in vacuo, while protected from light.
  • the recrystallization may be repeated more than once (e.g., 2, 3, 4, 5, or more times) to improve the impurity profile, e.g., until gossypol co-crystals comprise less than about 5%, 4%, 3%, 2%, 1%, 0.5%, or 0.1% impurities.
  • a final recrystallization may be carried out at a lower temperature, e.g., about -2O 0 C to about 1O 0 C, preferably about -1O 0 C to about 0 0 C.
  • the recrystallization mixture is held for about 15 minutes to about 100 minutes, e.g., about 30 to about 60 minutes, and the resulting crystals are then filtered, washed and dried as described above.
  • This method of producing gossypol acetic acid co-crystals produces a final product comprising less than about 5% total impurities, preferably less than about 3%, 2%, or 1%.
  • (-)-Gossypol acetic acid co-crystals may also be prepared from gossypol acetic acid co crystals via imine (Schiff base) formation with an optically active amine to form diastereomers.
  • the term "imine” includes other tautomers such as eneamine tautomers and stereoisomers thereof.
  • the optically active amine may be, but is not limited to, L-phenylalanine methyl ester, S-1-methylphenethylamine, or L- phenylalaninol or the corresponding HCl salt.
  • the derivatization may be carried out in the absence of oxygen, e.g., under a nitrogen purge.
  • the derivatization is carried out in the presence of a nonpolar and/or polar solvent, e.g., dichloromethane and/or isopropanol, for a time period of about 0.5 to about 3 hours, e.g., about 1 hour to about 2 hours.
  • a dehydrating agent such as sodium sulfate or a molecular sieve, e.g., type 3 A, is then added, along with suitable reagents for buffering the reaction mix at a pH of about 5 to about 7, e.g., about 6.
  • suitable reagents for buffering the reaction mix at a pH of about 5 to about 7, e.g., about 6.
  • One suitable buffering agent is sodium bicarbonate.
  • the reaction mixture is then stirred for at least about 15 minutes, e.g., at least about 30 minutes. The progression of the reaction may be monitored for completion. For example, the reaction mixture may be assayed for the absence of gossypol using thin layer chromatography (TLC) or preferably in real time by high pressure liquid chromatography (HPLC). If the reaction is incomplete, the pH may be adjusted back to about 6 by adding further buffering agents.
  • TLC thin layer chromatography
  • HPLC high pressure liquid chromatography
  • reaction is then continued for about 24 hours and again checked for completion of the reaction.
  • the reaction mixture may be filtered to remove the solids and the solids washed with additional non-polar solvent, e.g., dichloromethane.
  • the filtrates may then be evaporated to dryness, e.g., with a rotary evaporator with the bath set at about 3O 0 C to about 4O 0 C.
  • the resultant diastereomers are then separated, e.g., by chromatography
  • the diastereomers are separated by silica gel chromatography, e.g., Kromasil Si.
  • the dried filtrate is reconstituted in a non- polar solvent, e.g., dichloromethane, and charged onto the column.
  • the diastereomers are eluted with a solvent system comprising non-polar and polar solvents, e.g., 1 :1 heptane:ethyl acetate.
  • Column fractions may be monitored by HPLC and fractions containing the desired isomer (e.g., at least about 90%) may be pooled and evaporated.
  • Impure fractions may be collected and passed over the column additional times.
  • the diastereomer mixture is held for less than 24 hours, preferably less than 12, 6, or 3 hours, prior to separation in order to avoid any increase in contamination with gossypol derivatives.
  • the separated R-(-)-gossypol bis-imine diastereomer may then be hydrolyzed to produce (-)-gossypol.
  • the hydrolysis may be carried out in the absence of oxygen, e.g., under a nitrogen purge.
  • the (-)-gossypol derivative is mixed with a polar solvent (e.g., tetrahydrofuran) and an acid (e.g., aqueous hydrochloric acid) and stirred for at least about 1 hour, e.g., at least about 5 hours.
  • the extent of the reaction may be monitored by TLC or HPLC for depletion of both the bis- and mono-imine compounds to less than about 10%, preferably less than about 5%. If the reaction is not sufficient, it may be continued for at least about 15 hours and re-evaluated.
  • the reaction mixture may then be washed with an aqueous brine solution.
  • the aqueous brine solution may be back extracted with a polar solvent (e.g., ethyl acetate).
  • a polar solvent e.g., ethyl acetate
  • the organic layers are then combined and washed with an alkaline aqueous solution (e.g., sodium bicarbonate) followed by a brine solution.
  • the organic layers may then be evaporated to dryness, e.g., with a rotary evaporator with the bath set at about 3O 0 C to about 40 0 C.
  • the crude isolate is then dissolved in a solvent system comprising non- polar and polar solvents ⁇ e.g., 1 :1 heptane: ethyl acetate) and passed over a silica gel plug using the same solvent system containing a small amount of acetic acid (to avoid sticking to the plug).
  • a solvent system comprising non- polar and polar solvents ⁇ e.g., 1 :1 heptane: ethyl acetate
  • Fractions may be collected and monitored for gossypol content using TLC or HPLC.
  • Product-containing fractions may be pooled and evaporated to dryness, e.g., with a rotary evaporator with the bath set at about 3O 0 C to about 4O 0 C.
  • the (-)-gossypol may be purified by chromatography over a hydrophilic resin, e.g., a dihydroxypropyl resin such as DIOL, e.g., YMC DIOL (120 angstrom x 10-20 micron) (GL Sciences).
  • a hydrophilic resin e.g., a dihydroxypropyl resin such as DIOL, e.g., YMC DIOL (120 angstrom x 10-20 micron) (GL Sciences).
  • DIOL dihydroxypropyl resin
  • YMC DIOL 120 angstrom x 10-20 micron
  • the dried product from the previous step may be reconstituted in a solvent system comprising non-polar and polar solvents (e.g., 1 :1 heptane:ethyl acetate) and purified over the column using the same solvent system.
  • Fractions are collected, held at a reduced temperature (e.g., about 2°C to about 8°C), and the fractions assayed for gossypol content using TLC or HPLC.
  • Fractions containing gossypol e.g., at least 90%
  • Fractions with less than 90% gossypol may be pooled and re-purified over the column.
  • the purified (-)-gossypol may be dissolved in acetone
  • the mixture may then be loaded into a suitable container for crystallization (e.g., a B ⁇ chi Ball). If there is no immediate crystallization, the solvent may be slowly removed by vacuum until a crystal mass appears. The mixture may then be held for about 15 minutes to about 100 minutes, e.g., about 30 minutes to about 60 minutes, and then filtered. The crystals may then be washed with the same ratio of acetone and acetic acid.
  • a suitable container for crystallization e.g., a B ⁇ chi Ball
  • the solvent may be slowly removed by vacuum until a crystal mass appears.
  • the mixture may then be held for about 15 minutes to about 100 minutes, e.g., about 30 minutes to about 60 minutes, and then filtered.
  • the crystals may then be washed with the same ratio of acetone and acetic acid.
  • the crystals may be soaked in acetic acid (about 3 mL per 1 g gossypol for about 20 to about 40 minutes, preferably about 30 minutes, and the acetic acid removed by filtration.
  • the crystals may then be dried (e.g. , in vacuo) for at least one hour, e.g., about 2 to about 4 hours.
  • the crystals may be packaged and stored protected from light (e.g., in amber glass vials) at a reduced temperature (e.g., about -3O 0 C to about 0 0 C, preferably about -10 0 C to about -2O 0 C.
  • An alternative method of derivatizing gossypol acetic acid starting material comprising ( ⁇ )-gossypol to form a Schiff base in the above-described methods of producing (-)-gossypol acetic acid co-crystals comprises treating a mixture of gossypol acetic acid starting material and optically active amine (e.g., L-phenylalanine methyl ester hydrochloride) in a nonpolar solvent (e.g., dichloromethane) with triethylamine and mixing for at least 2 hours, e.g., about 5 hours, optionally under an oxygen-free atmosphere.
  • the reaction may be monitored for completion using HPLC or TLC methods. After completion of the reaction, the mixture is extracted with water and the organic phase separated and evaporated to dryness, e.g., with a rotary evaporator with the bath set at about 25°C to about 35°C, followed by a high vacuum overnight.
  • gossypol been shown to bind to Bcl-2 and BCI-X L at the BH3 binding groove and to have anticancer activity (U.S. Patent Application No. 2003/0008924).
  • gossypol may be used to induce apoptosis and also potentiate the induction of apoptosis in response to apoptosis induction signals when administered by pulsatile dosing. It is contemplated that gossypol administered by pulsatile dosing will sensitize cells to inducers of apoptosis, including cells that are resistant to such inducers.
  • Gossypol administered by pulsatile dosing can be used to induce apoptosis in any disorder that can be treated, ameliorated, or prevented by the induction of apoptosis.
  • the present invention provides methods for targeting patients characterized as overexpressing an anti-apoptotic Bcl-2 family protein.
  • the cells e.g., cancer cells
  • the cells operationally manifest elevated expression levels of anti-apoptotic Bcl-2 family proteins by virtue of executing the apoptosis program and dying in response to administration of an inhibiting effective amount of gossypol, said response occurring, at least in part, due to the dependence in such cells on anti-apoptotic Bcl-2 family protein function for their survival.
  • the methods of the present invention are used to treat diseased cells, tissues, organs, or pathological conditions and/or disease states in a patient (e.g., a mammalian subject including, but not limited to, humans and veterinary animals).
  • a patient e.g., a mammalian subject including, but not limited to, humans and veterinary animals.
  • various diseases and pathologies are amenable to treatment or prophylaxis using the present methods.
  • a non-limiting exemplary list of these diseases and conditions includes, but is not limited to, cancers, T and B cell mediated autoimmune diseases, inflammatory diseases, infections, hyperproliferative diseases, AIDS, degenerative conditions, vascular diseases, and the like.
  • the cancer cells being treated are metastatic.
  • the cancer cells being treated are resistant to anticancer agents, e.g., taxanes, e.g., docetaxel.
  • infections suitable for treatment with the methods of the present invention include, but are not limited to, infections caused by viruses, bacteria, fungi, parasites, mycoplasma, prions, and the like.
  • gossypol administered by pulsatile doing can be used to modulate spermicidal activity (e.g., function as a male contraceptive or antifertility agent), treat malaria, microbial or viral disease (e.g., inhibit the growth of the HIV virus as a treatment for AIDS), treat obesity, skin disorders or baldness, inhibit growth of endothelial cells, inhibit vascularization or neovascularization, treat arthritic conditions, neovascular-based dermatological conditions, diabetic retinopathy, Kaposi's sarcoma, age-related macular degeneration, restenosis, telangiectasia, glaucoma, keloids, corneal graft rejection, wound granularization, angiofibroma, Osier- Webber syndrome, myocardial angiogenesis, p
  • Some embodiments of the present invention provides methods for administering an effective amount of gossypol by pulsatile dosing and at least one additional therapeutic agent (including, but not limited to, chemotherapeutic agents, antineoplastic agents, antimicrobial agents, antiviral agents, antifungal agents, and anti-inflammatory agents) and/or therapeutic technique (e.g., surgical intervention, and/or radiotherapeutic agent).
  • additional therapeutic agent including, but not limited to, chemotherapeutic agents, antineoplastic agents, antimicrobial agents, antiviral agents, antifungal agents, and anti-inflammatory agents
  • therapeutic technique e.g., surgical intervention, and/or radiotherapeutic agent.
  • chemotherapeutic agent refers to any chemical substance known to those of skill in the art to be effective for the treatment or amelioration of cancer and/or as an inducer of apoptosis.
  • the combination of gossypol administered by pulsatile dosing and one or more therapeutic agents will have a greater effect as compared to the administration of either gossypol or therapeutic agent alone.
  • the combination of gossypol administered by pulsatile dosing and one or more therapeutic agents is expected to result in a synergistic effect (i.e., more than additive) as compared to the administration of either one alone.
  • pulsatile dose administration of gossypol results in a reduction in number and/or decreased severity of adverse events in patients.
  • the combination of gossypol administered by pulsatile dosing and one or more anticancer agents overcomes patient chemoresistance to the anticancer agent.
  • the invention pertains to a method of treating, ameliorating or preventing cancer comprising administering to a patient in need thereof (-)-gossypol in combination with an anticancer agent, wherein about 40 mg to about 60 mg of said (-)-gossypol is administered twice-a-day for three consecutive days followed by eighteen consecutive days wherein said (-)-gossypol is not administered.
  • the anticancer agent is docetaxel.
  • the patient has demonstrated chemoresi stance to docetaxel treatment.
  • the cancer is prostate cancer.
  • anticancer agents are contemplated for use in the methods of the present invention. Indeed, the present invention contemplates, but is not limited to, administration of numerous anticancer agents such as: agents that induce apoptosis; polynucleotides (e.g., anti-sense, ribozymes, siRNA); polypeptides (e.g., enzymes and antibodies); biological mimetics (e.g., BH3 mimetics); agents that bind (e.g., oligomerize or complex) with a Bcl-2 family protein such as Bax; alkaloids; alkylating agents; antitumor antibiotics; antimetabolites; hormones; platinum compounds; monoclonal or polyclonal antibodies (e.g., antibodies conjugated with anticancer drugs, toxins, defensins), toxins; radionuclides; biological response modifiers (e.g., interferons (e.g., IFN- ⁇ ) and interleukins (e
  • anticancer agents comprise agents that induce or stimulate apoptosis.
  • Agents that induce apoptosis include, but are not limited to, radiation (e.g., X-rays, gamma rays, UV); kinase inhibitors (e.g., epidermal growth factor receptor (EGFR) kinase inhibitor, vascular growth factor receptor (VGFR) kinase inhibitor, fibroblast growth factor receptor (FGFR) kinase inhibitor, platelet-derived growth factor receptor (PDGFR) kinase inhibitor, and Bcr-Abl kinase inhibitors (such as GLEEVEC)); antisense molecules; antibodies (e.g., HERCEPTIN, RITUXAN, ZEVALIN, BEXXAR, and AVASTIN); anti-estrogens (e.g., raloxifene and tamoxifen); anti-androgens (e.g., flutamide, b
  • the methods of the present invention provide pulsatile dose administration of gossypol and at least one anti- hyperproliferative or antineoplastic agent; e.g., selected from alkylating agents, antimetabolites, and natural products (e.g., herbs and other plant and/or animal derived compounds).
  • at least one anti- hyperproliferative or antineoplastic agent e.g., selected from alkylating agents, antimetabolites, and natural products (e.g., herbs and other plant and/or animal derived compounds).
  • Alkylating agents suitable for use in the present compositions and methods include, but are not limited to: 1) nitrogen mustards (e.g., mechlorethamine, cyclophosphamide, ifosfamide, melphalan (L-sarcolysin); and chlorambucil); 2) ethylenimines and methylmelamines (e.g., hexamethylmelamine and thiotepa); 3) alkyl sulfonates (e.g., busulfan); 4) nitrosoureas (e.g., carmustine (BCNU); lomustine (CCNU); semustine (methyl-CCNU); and streptozocin (streptozotocin)); and 5) triazenes (e.g., dacarbazine (dimethyltriazenoimid-azolecarboxamide) .
  • nitrogen mustards e.g., mechlorethamine, cyclophosphamide,
  • antimetabolites suitable for use in the present compositions and methods include, but are not limited to: 1) folic acid analogs (e.g., methotrexate (amethopterin)); 2) pyrimidine analogs (e.g., fluorouracil (5-fluorouracil), floxuridine (fluorode-oxyuridine), and cytarabine (cytosine arabinoside)); and 3) purine analogs (e.g., mercaptopurine (6-mercaptopurine), thioguanine (6-thioguanine), and pentostatin (2'-deoxycoformycin)).
  • folic acid analogs e.g., methotrexate (amethopterin)
  • pyrimidine analogs e.g., fluorouracil (5-fluorouracil), floxuridine (fluorode-oxyuridine), and cytarabine (cytosine arabinoside)
  • purine analogs e.g
  • chemotherapeutic agents suitable for use in the compositions and methods of the present invention include, but are not limited to: 1) vinca alkaloids (e.g., vinblastine, vincristine); 2) epipodophyllotoxins (e.g., etoposide and teniposide); 3) antibiotics (e.g., dactinomycin (actinomycin D), daunorubicin (daunomycin; rubidomycin), doxorubicin, bleomycin, plicamycin (mithramycin), and mitomycin (mitomycin C)); 4) enzymes (e.g., L-asparaginase); 5) biological response modifiers (e.g., interferon-alfa); 6) platinum coordinating complexes (e.g., cisplatin and carboplatin); 7) anthracenediones (e.g., mitoxantrone); 8) substituted ureas (
  • any oncolytic agent that is used in a cancer therapy context finds use in the methods of the present invention.
  • the U.S. Food and Drug Administration maintains a formulary of oncolytic agents approved for use in the United States. International counterpart agencies to the U.S.F.D.A. maintain similar formularies.
  • Table 1 provides a list of exemplary antineoplastic agents approved for use in the U.S. Those skilled in the art will appreciate that the "product labels" required on all U.S. approved chemotherapeutics describe approved indications, dosing information, toxicity data, and the like, for the exemplary agents.
  • Table 1 provides a list of exemplary antineoplastic agents approved for use in the U.S. Those skilled in the art will appreciate that the "product labels" required on all U.S. approved chemotherapeutics describe approved indications, dosing information, toxicity data, and the like, for the exemplary agents.
  • Anticancer agents further include compounds which have been identified to have anticancer activity but are not currently approved by the U.S. Food and Drug Administration or other counterpart agencies or are undergoing evaluation for new uses. Examples include, but are not limited to, 3-AP, 12-O-tetradecanoylphorbol-13-acetate, 17AAG, 852 A, ABI-007, ABR- 217620, ABT-751, ADI-PEG 20, AE-941, AG-013736, AGROlOO, alanosine, AMG 706, antibody G250, antineoplastons, AP23573, apaziquone, APC8015, atiprimod, ATN-161, atrasenten, azacitidine, BB-10901, BCX-1777, bevacizumab, BGOOOOl, bicalutamide, BMS 247550, bortezomib, bryostatin- 1, buserelin, calcitriol, CCI-779, CDB-2914, cefixime,
  • the anticancer agent is selected from the group consisting of abraxane, actinomycin D, aldesleukin, alemtuzumab, alitretinoin, allopurinol, altretamine, amifostine, aminoglutethamide, anastrozole, arsenic trioxide, asparaginase, azacitidine, azathioprine, BCG live, bevacizumab, bexarotene, bicalutamide, bleomycin, bortezomib, busulfan, butazolidin, capecitabine, carboplatin, carmustine, celecoxib, chlorambucil, cisplatin, cladribine, clofarabine, cyclophosphamide, cytarabine, dacarbazine, dactinomycin, darbepoetin alfa, daunomycin, daunorubicin, denile
  • the anticancer agent is a taxane.
  • the taxane is selected from the group consisting of docetaxel and paclitaxel. In another embodiment, the taxane is docetaxel.
  • the present invention provides methods for pulsatile dose administration of gossypol with radiation therapy.
  • radiation therapy refers any type of radiation therapy known to those of skill in the art to be effective for the treatment or amelioration of cancer and/or as an inducer of apoptosis.
  • the invention is not limited by the types, amounts, or delivery and administration systems used to deliver the therapeutic dose of radiation to the patient.
  • the patient may receive photon radiotherapy, particle beam radiation therapy, radioisotope therapy (e.g., radioconjugates with monoclonal antibodies), other types of radiotherapies, and combinations thereof.
  • the radiation is delivered to the patient using a linear accelerator.
  • the radiation is delivered using a gamma knife.
  • the source of radiation can be external or internal to the patient.
  • External radiation therapy is most common and involves directing a beam of high-energy radiation to a tumor site through the skin using, for instance, a linear accelerator. While the beam of radiation is localized to the tumor site, it is nearly impossible to avoid exposure of normal, healthy tissue. However, external radiation is usually well tolerated by patients.
  • Internal radiation therapy involves implanting a radiation-emitting source, such as beads, wires, pellets, capsules, particles, and the like, inside the body at or near the tumor site including the use of delivery systems that specifically target cancer cells (e.g., using particles attached to cancer cell binding ligands). Such implants can be removed following treatment, or left in the body inactive.
  • Types of internal radiation therapy include, but are not limited to, brachytherapy, interstitial irradiation, intracavity irradiation, radioimmunotherapy, and the like.
  • the patient may optionally receive radiosensitizers (e.g., metronidazole, misonidazole, intra-arterial Budr, intravenous iododeoxyuridine (IudR), nitroimidazole, 5-substituted-4-nitroimidazoles, 2H- isoindolediones, [[(2-bromoethyl)-amino]methyl]-nitro-lH-imidazole-l- ethanol, nitroaniline derivatives, DNA-aff ⁇ nic hypoxia selective cytotoxins, halogenated DNA ligand, 1,2,4 benzotriazine oxides, 2-nitroimidazole derivatives, fluorine-containing nitroazole derivatives, benzamide, nicotinamide, acridine-intercalator, 5-thiotretrazole derivative, 3-nitro-l,2,4- triazole, 4,5-dinitroimidazole derivative, hydroxylated texaph
  • any type of radiation can be administered to a patient, so long as the dose of radiation is tolerated by the patient without unacceptable negative side-effects.
  • Suitable types of radiotherapy include, for example, ionizing (electromagnetic) radiotherapy (e.g., X-rays or gamma rays) or particle beam radiation therapy (e.g., high linear energy radiation).
  • Ionizing radiation is defined as radiation comprising particles or photons that have sufficient energy to produce ionization, i.e., gain or loss of electrons (as described in, for example, U.S. 5,770,581 incorporated herein by reference in its entirety).
  • the effects of radiation can be at least partially controlled by the clinician.
  • the dose of radiation is preferably fractionated for maximal target cell exposure and reduced toxicity.
  • the total dose of radiation administered to an animal preferably is about .01 Gray (Gy) to about 100 Gy. More preferably, about 10 Gy to about 65 Gy (e.g., about 15 Gy, 20 Gy, 25 Gy, 30 Gy, 35 Gy, 40 Gy, 45 Gy, 50 Gy, 55 Gy, or 60 Gy) are administered over the course of treatment. While in some embodiments a complete dose of radiation can be administered over the course of one day, the total dose is ideally fractionated and administered over several days. Desirably, radiotherapy is administered over the course of at least about 3 days, e.g., at least 5, 7, 10, 14, 17, 21, 25, 28, 32, 35, 38, 42, 46, 52, or 56 days (about 1-8 weeks).
  • a daily dose of radiation will comprise approximately 1-5 Gy (e.g., about 1 Gy, 1.5 Gy, 1.8 Gy, 2 Gy, 2.5 Gy, 2.8 Gy, 3 Gy, 3.2 Gy, 3.5 Gy, 3.8 Gy, 4 Gy, 4.2 Gy, or 4.5 Gy), preferably 1-2 Gy (e.g., 1.5-2 Gy).
  • the daily dose of radiation should be sufficient to induce destruction of the targeted cells.
  • radiation preferably is not administered every day, thereby allowing the animal to rest and the effects of the therapy to be realized.
  • radiation desirably is administered on 5 consecutive days, and not administered on 2 days, for each week of treatment, thereby allowing 2 days of rest per week.
  • radiation can be administered 1 day/week, 2 days/week, 3 days/week, 4 days/week, 5 days/week, 6 days/week, or all 7 days/week, depending on the animal's responsiveness and any potential side effects.
  • Radiation therapy can be initiated at any time in the therapeutic period.
  • radiation is initiated in week 1 or week 2, and is administered for the remaining duration of the therapeutic period.
  • radiation is administered in weeks 1-6 or in weeks 2-6 of a therapeutic period comprising 6 weeks for treating, for instance, a solid tumor.
  • radiation is administered in weeks 1-5 or weeks 2-5 of a therapeutic period comprising 5 weeks.
  • Antimicrobial therapeutic agents may also be used as therapeutic agents in the present invention. Any agent that can kill, inhibit, or otherwise attenuate the function of microbial organisms may be used, as well as any agent contemplated to have such activities.
  • Antimicrobial agents include, but are not limited to, natural and synthetic antibiotics, antibodies, inhibitory proteins (e.g., defensins), antisense nucleic acids, membrane disruptive agents and the like, used alone or in combination. Indeed, any type of antibiotic may be used including, but not limited to, antibacterial agents, antiviral agents, antifungal agents, and the like.
  • gossypol and one or more therapeutic agents are administered to an animal under one or more of the following conditions: at different periodicities, at different durations, at different concentrations, by different administration routes, etc. provided that gossypol is administered according to a discontinuous dosing regimen (i.e., via pulsatile dosing).
  • gossypol is administered by pulsatile dosing prior to the therapeutic agent, e.g., 0.5, 1, 2, 3, 4, 5, 10, 12, or 18 hours, 1, 2, 3, 4, 5, or 6 days, 1, 2, 3, or 4 weeks prior to the administration of the therapeutic agent.
  • gossypol is administered by pulsatile dosing after the therapeutic agent, e.g., 0.5, 1, 2 3, 4, 5, 10, 12, or 18 hours, 1, 2, 3, 4, 5, or 6 days, 1, 2, 3, or 4 weeks after the administration of the therapeutic agent.
  • gossypol and the therapeutic agent are administered concurrently but on different schedules, e.g., gossypol is administered on at least two consecutive days followed by at least one day wherein gossypol is not administered while the therapeutic agent is administered once a week, once every two weeks, once every three weeks, or once every four weeks.
  • gossypol is administered on one day a week while the therapeutic agent is administered daily, once a week, once every two weeks, once every three weeks, or once every four weeks.
  • (-)-gossypol is administered twice-a-day for three consecutive days every 21 days, one therapeutic agent, e.g., docetaxel, is administered every 21 days, and another therapeutic agent, e.g., prednisone, is administered daily.
  • compositions may be produced by combining gossypol in a therapeutically effective amount to induce apoptosis in cells or to sensitize cells to inducers of apoptosis with a pharmaceutically acceptable carrier.
  • Pharmaceutical compositions may comprise, for example, ( ⁇ )-gossypol, (+)- gossypol, (-)-gossypol, ( ⁇ )-gossypol co-crystal, (+)-gossypol co-crystal or (-)- gossypol co-crystal.
  • compositions useful within the scope of this invention include all compositions wherein gossypol is contained in an amount which is effective to achieve its intended purpose, e.g., to treat, ameliorate or prevent a disease, condition or disorder responsive to the induction of apoptosis. While individual needs vary, determination of optimal ranges of effective amounts of each component in a pharmaceutical composition is within the skill of the art.
  • the pharmaceutical composition comprising gossypol may be administered to patients, e.g. humans, orally at a dose totaling about 1 mg to about 1200 mg, or an equivalent dose of the pharmaceutically acceptable salt thereof, per day.
  • a total oral dose of about 5 mg to about 500 mg, about 5 mg to about 250 mg, about 5 mg to about 100 mg, or about 5 mg to about 60 mg is administered per day.
  • a total oral dose of about 90 mg to about 240 mg is administered per day.
  • a total oral dose of about 80 mg to about 200 mg is administered per day.
  • an oral dose of about 5 mg, 10 mg, 20 mg, 30 mg, 40 mg, 50 mg, 60 mg, 70 mg, 80 mg, 90 mg, 100 mg, 110 mg, 120 mg, 130 mg, 140 mg, 150 mg, 160 mg, 170 mg, 180 mg, 190 mg or 200 mg is administered two times a day.
  • an oral dose of 30 mg, 35 mg, 40 mg, 45 mg, 50 mg, 55 mg, 60 mg, 65 mg, 70 mg, 75 mg or 80 mg is administered two times a day.
  • an oral dose of 80 mg, 90 mg, 100 mg, 110 mg, 120 mg, 130 mg, 140 mg, 150 mg, 160 mg, 170 mg, 180 mg, 190 mg or 200 mg is administered once a day.
  • the amount (e.g., dose) of gossypol administered may increase as the period of time between dosing increases, since the potential for adverse events may decrease under such circumstances.
  • the unit oral dose may comprise from about 1 mg to about 1000 mg of the pharmaceutical composition comprising gossypol. In one embodiment, the unit oral dose may comprise from about 5 mg to about 500 mg of the pharmaceutical composition comprising gossypol. In another embodiment, the unit oral dose may comprise from about 5 mg to about 100 mg.
  • the unit oral dose may comprise from about 5 mg to about 30 mg. In another embodiment, the unit oral dose may comprise from about 30 mg to about 80 mg. In another embodiment, the unit oral dose may comprise about 40 mg to about 60 mg.
  • the unit dose may be administered one or more times daily as one or more tablets, capsules and the like, each containing from about 1 to about 1000 mg of the pharmaceutical composition comprising gossypol, conveniently about 5 mg to about 100 mg of the composition, e.g., about 1 mg, 5 mg, 10 mg, 15 mg, 20 mg, 25 mg, 30 mg, 35 mg, 40 mg, 45 mg, 50 mg, 55 mg, 60 mg, 65 mg, 70 mg, 75 mg, 80 mg, 85 mg, 90 mg, 95 mg or 100 mg.
  • the unit oral dose is administered two times per day. In another embodiment, the unit oral dose is administered two times per day for three consecutive days.
  • the dose is generally about one-half of the oral dose.
  • a suitable intramuscular dose of gossypol would be about 0.5 mg to about 500 mg.
  • the intramuscular dose would be about 0.5 mg to about 100 mg, about 0.5 to about 50 mg, about 0.5 mg to about 25 mg, or about 0.5 mg to about 15 mg.
  • the composition may be present at a concentration of about 0.01 to 100 mg per gram of carrier. In one embodiment, the composition is present at a concentration of about 0.07 mg/g to about 1.0 mg/g, about 0.1 to about 0.5 mg/g, or about 0.4 mg/g.
  • Gossypol may be administered via pulsatile dosing as part of a pharmaceutical composition containing suitable pharmaceutically acceptable carriers comprising excipients and auxiliaries which facilitate processing of the compositions into preparations which can be used pharmaceutically.
  • Pharmaceutical preparations which can be administered orally include tablets, dragees, slow release lozenges, capsules and the like.
  • Pharmaceutical preparations which can be administered topically include mouth rinses and mouth washes, gels, liquid suspensions, hair rinses, hair gels, shampoos and the like.
  • Pharmaceutical preparations which can be administered rectally include suppositories and the like.
  • Pharmaceutical preparations for administration by injection include suitable solutions and the like.
  • compositions administered via pulsatile dosing contain from about 0.01 to about 99 percent, or from about 0.25 to about 75 percent of gossypol, together with the excipient(s).
  • Methods of the invention may be administered to any patient which may experience the beneficial effects of such methods. Foremost among such patients are mammals, e.g., humans, although the invention is not intended to be so limited. Other patients include veterinary animals (cows, sheep, pigs, horses, dogs, cats and the like).
  • Gossypol and pharmaceutical compositions thereof may be administered via pulsatile dose administration by any means that achieve their intended purpose.
  • administration may be by parenteral, subcutaneous, intravenous, intramuscular, intraperitoneal, transdermal, buccal, intrathecal, intracranial, intranasal or topical routes.
  • administration may be by the oral route.
  • the dosage administered will be dependent upon the age, health, and weight of the recipient, kind of concurrent treatment, if any, frequency of treatment, and the nature of the effect desired.
  • compositions of the present invention are manufactured in a manner which is itself known, for example, by means of conventional mixing, granulating, dragee-making, dissolving, or lyophilizing processes.
  • pharmaceutical preparations for oral use can be obtained by combining the active compounds with solid excipients, optionally grinding the resulting mixture and processing the mixture of granules, after adding suitable auxiliaries, if desired or necessary, to obtain tablets or dragee cores.
  • Suitable excipients are, in particular, fillers such as saccharides, for example lactose or sucrose, mannitol or sorbitol, cellulose preparations and/or calcium phosphates, for example tricalcium phosphate or calcium hydrogen phosphate, as well as binders such as starch paste, using, for example, maize starch, wheat starch, rice starch, potato starch, gelatin, tragacanth, methyl cellulose, hydroxypropylmethylcellulose, sodium carboxymethylcellulose, and/or polyvinyl pyrrolidone.
  • fillers such as saccharides, for example lactose or sucrose, mannitol or sorbitol, cellulose preparations and/or calcium phosphates, for example tricalcium phosphate or calcium hydrogen phosphate, as well as binders such as starch paste, using, for example, maize starch, wheat starch, rice starch, potato starch, gelatin, tragacanth, methyl cellulose,
  • disintegrating agents may be added such as the above-mentioned starches and also carboxymethyl-starch, cross- linked polyvinyl pyrrolidone, agar, or alginic acid or a salt thereof, such as sodium alginate.
  • Auxiliaries are, above all, flow-regulating agents and lubricants, for example, silica, talc, stearic acid or salts thereof, such as magnesium stearate or calcium stearate, and/or polyethylene glycol.
  • Dragee cores are provided with suitable coatings which, if desired, are resistant to gastric juices.
  • concentrated saccharide solutions may be used, which may optionally contain gum arabic, talc, polyvinyl pyrrolidone, polyethylene glycol and/or titanium dioxide, lacquer solutions and suitable organic solvents or solvent mixtures.
  • suitable cellulose preparations such as acetylcellulose phthalate or hydroxypropylmethyl-cellulose phthalate, are used.
  • Dye stuffs or pigments may be added to the tablets or dragee coatings, for example, for identification or in order to characterize combinations of active compound doses.
  • Other pharmaceutical preparations which can be used orally include push-fit capsules made of gelatin, as well as soft, sealed capsules made of gelatin and a plasticizer such as glycerol or sorbitol.
  • the push-fit capsules can contain the active compounds in the form of granules which may be mixed with fillers such as lactose, binders such as starches, and/or lubricants such as talc or magnesium stearate and, optionally, stabilizers.
  • the active compounds are preferably dissolved or suspended in suitable liquids, such as fatty oils, or liquid paraffin.
  • stabilizers may be added.
  • Possible pharmaceutical preparations which can be used rectally include, for example, suppositories, which consist of a combination of one or more of the active compounds with a suppository base.
  • Suitable suppository bases are, for example, natural or synthetic triglycerides, or paraffin hydrocarbons.
  • gelatin rectal capsules which consist of a combination of the active compounds with a base.
  • Possible base materials include, for example, liquid triglycerides, polyethylene glycols, or paraffin hydrocarbons.
  • Suitable formulations for parenteral administration include aqueous solutions of the active compounds in water-soluble form, for example, water- soluble salts and alkaline solutions.
  • suspensions of the active compounds as appropriate oily injection suspensions may be administered.
  • Suitable lipophilic solvents or vehicles include fatty oils, for example, sesame oil, or synthetic fatty acid esters, for example, ethyl oleate or triglycerides or polyethylene glycol-400.
  • Aqueous injection suspensions may contain substances which increase the viscosity of the suspension include, for example, sodium carboxymethyl cellulose, sorbitol, and/or dextran.
  • the suspension may also contain stabilizers.
  • the topical compositions of this invention are formulated preferably as oils, creams, lotions, ointments and the like by choice of appropriate carriers.
  • Suitable carriers include vegetable or mineral oils, white petrolatum (white soft paraffin), branched chain fats or oils, animal fats and high molecular weight alcohol (greater than C 12 ).
  • the preferred carriers are those in which the active ingredient is soluble.
  • Emulsifiers, stabilizers, humectants and antioxidants may also be included as well as agents imparting color or fragrance, if desired.
  • transdermal penetration enhancers can be employed in these topical formulations. Examples of such enhancers can be found in U.S. Pat. Nos. 3,989,816 and 4,444,762.
  • Creams are preferably formulated from a mixture of mineral oil, self- emulsifying beeswax and water in which mixture the active ingredient, dissolved in a small amount of an oil such as almond oil, is admixed.
  • a typical example of such a cream is one which includes about 40 parts water, about 20 parts beeswax, about 40 parts mineral oil and about 1 part almond oil.
  • Ointments may be formulated by mixing a suspension of the active ingredient in a vegetable oil such as almond oil with warm soft paraffin and allowing the mixture to cool.
  • a vegetable oil such as almond oil
  • a typical example of such an ointment is one which includes about 30% almond oil and about 70% white soft paraffin by weight.
  • Lotions may be conveniently prepared by preparing a suspension of the active ingredient in a suitable high molecular weight alcohol such as propylene glycol or polyethylene glycol.
  • a suitable high molecular weight alcohol such as propylene glycol or polyethylene glycol.
  • a phase I clinical trial was carried out to compare the maximum tolerated dose and safety of daily (i.e., continuous) versus pulsatile (i.e., intermittent) dosing of (-)-gossypol in patients with advanced cancer.
  • a secondary objective of this study was to identify any anti-tumor activity of (-)-gossypol.
  • (-)-gossypol acetic acid co- crystals were administered at 60 mg/kg, po, daily for three days per week (day 1-3/week) every two weeks (days 1-3, and then days 15-17), either alone or in combination with taxotere at 30 mg/kg, iv, single dose only, once every three weeks (Fig. 3).
  • the results of these studies show inter alia that an intermittent dosing of (-)-gossypol acetic acid co-crystals in combination with taxotere effectively reduces tumor volume.
  • Phase I data from 9 patients is summarized as follows: no dose- limiting toxicities, no severe gastrointestinal toxicity, no increase in docetaxel hematological toxicity.
  • Phase I-II data is available on first 20 patients of Cohort A.
  • the patient demographics are provided in Table 4. See N. Engl. J. Med. 357:1502- 1512 (2004) for further details of TAX 327 clinical study.
  • Figure 4 is a waterfall plot illustrating the therapeutic response of the first 20 patients of Cohort A. These data indicate a high rate of PSA response with (-)-gossypol acetic acid co-crystal treatment. In addition, only one patient out of 20 was refractory at the onset of treatment Data from this study also indicates steep declines in the slope of PSA response versus time and PSA reductions occur early in the treatment and are long lasting (Figure 5). Finally, CT scanning shows tumor size reduction following treatment with (-)- gossypol, docetaxel, and prednisone (Figure 6). Table 5 shows the preliminary efficacy and safety comparisons between CS-202 and TAX 327.
  • Table 6 compares the PSA responses of (-)-gossypol acetic acid co- crystal Cohort A combination therapy with approved docetaxel monotherapy regimens and other investigational combination therapies for the treatment of hormone refractory prostate cancer.
  • (-)-Gossypol acetic acid co-crystal (labeled "(-)-gossypol”) responses compare favorably to those reported in these other studies.
  • Figure 7 illustrates the therapeutic response of the first 11 patients of
  • (-)-gossypol acetic acid co-crystal is added to a taxotere regimen of various patients of Cohort B.
  • the abrupt reversal of the PSA response suggests pulsatile dosing of (-)-gossypol acetic acid co-crystal may be especially effective in treating chemoresistant patients.

Landscapes

  • Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Organic Chemistry (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Epidemiology (AREA)
  • Immunology (AREA)
  • Urology & Nephrology (AREA)
  • Oncology (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Communicable Diseases (AREA)
  • Dermatology (AREA)
  • Rheumatology (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Neurology (AREA)
  • Transplantation (AREA)
  • Pain & Pain Management (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Biomedical Technology (AREA)
  • Cardiology (AREA)
  • Neurosurgery (AREA)
  • Vascular Medicine (AREA)
  • Hematology (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Virology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Pulmonology (AREA)
  • Endocrinology (AREA)
  • Reproductive Health (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

This invention relates to pulsatile dose administration of gossypol or pharmaceutical compositions thereof for treating diseases, disorders and conditions responsive to gossypol, inhibiting the activity of anti-apoptotic Bcl-2 family proteins, inducing apoptosis in cells and increasing the sensitivity of cells to inducers of apoptosis.

Description

PULSATILE DOSING OF GOSSYPOL FOR TREATMENT OF DISEASE
BACKGROUND OF THE INVENTION
Field of the Invention
[0001] This invention is in the field of medicinal chemistry. In particular, the invention relates to pulsatile dose administration of gossypol or pharmaceutical compositions thereof for treating diseases, disorders and conditions responsive to gossypol, inhibiting the activity of anti-apoptotic Bcl-2 family proteins, inducing apoptosis in cells and increasing the sensitivity of cells to inducers of apoptosis.
Related Art
[0002] The aggressive cancer cell phenotype is the result of a variety of genetic and epigenetic alterations leading to deregulation of intracellular signaling pathways (Ponder, Nature 411:336 (2001)). The commonality for all cancer cells, however, is their failure to execute an apoptotic program, and lack of appropriate apoptosis due to defects in the normal apoptosis machinery is a hallmark of cancer (Lowe et ai, Carcinogenesis 27:485 (2000)). Most of the current cancer therapies, including chemotherapeutic agents, radiation (radiotherapeutic agents), and immunotherapy, work by indirectly inducing apoptosis in cancer cells. The inability of cancer cells to execute an apoptotic program due to defects in the normal apoptotic machinery is thus often associated with an increase in resistance to chemotherapy, radiation, or immunotherapy-induced apoptosis. Primary or acquired resistance of human cancer of different origins to current treatment protocols due to apoptosis defects is a major problem in current cancer therapy (Lowe et al, Carcinogenesis 27:485 (2000); Nicholson, Nature 407:810 (2000)). Accordingly, current and future efforts towards designing and developing new molecular target-specific anticancer therapies to improve survival and quality of life of cancer patients must include strategies that specifically target cancer cell resistance to apoptosis. In this regard, targeting crucial negative regulators that play a central role in directly inhibiting apoptosis in cancer cells represents a highly promising therapeutic strategy for new anticancer drug design.
[0003] Two classes of central negative regulators of apoptosis have been identified. The first class of regulators is the inhibitor of apoptosis proteins (IAPs) (Deveraux et al, Genes Dev. 13:239 (1999); Salvesen et al, Nat. Rev. MoI. Cell. Biol. 5:401 (2002)). IAP proteins potently suppress apoptosis induced by a large variety of apoptotic stimuli, including chemotherapeutic agents, radiation, and immunotherapy in cancer cells.
[0004] The second class of central negative regulators of apoptosis is the BcI-
2 family of proteins (Adams et al, Science 281:1322 (1998); Reed, Adv. Pharmacol. 41:501 (1997); Reed et al, J. Cell. Biochem. 60:23 (1996)). Bcl-2 is the founding member of the family and was first isolated as the product of an oncogene. The Bcl-2 family now includes both anti-apoptotic molecules such as Bcl-2 and BCI-XL and pro-apoptotic molecules such as Bax, Bak, Bid, and Bad. Bcl-2 and BCI-XL are overexpressed in many types of human cancer {e.g., breast, prostate, colorectal, lung, etc.), including Non-Hodgkin's lymphoma, which is caused by a chromosomal translocation (tl4, 18) that leads to overexpression of Bcl-2. This suggests that many cancer cell types depend on the elevated levels of Bcl-2 and/or BCI-XL to survive the other cellular derangements that simultaneously both define them as cancerous or pre-cancerous cells and cause them to attempt to execute the apoptosis pathway. Also, increased expression of Bcl-2 family proteins has been recognized as a basis for the development of resistance to cancer therapeutic drugs and radiation that act in various ways to induce cell death in tumor cells.
[0005] Bcl-2 and BCI-XL are thought to play a role in tumor cell migration and invasion, and therefore, metastasis (Amberger et al., Cancer Res. 55:149 (1998); Wick et al, FEBS Lett. 440:419 (1998); Mohanam et al., Cancer Res. 53:4143 (1993); Pedersen et al, Cancer Res. 55:5158 (1993)). Bcl-2 family proteins appear to provide tumor cells with a mechanism for surviving in new and non-permissive environments (e.g., metastatic sites), and contribute to the organospecific pattern of clinical metastatic cancer spread (Rubio, Lab Invest. 81:725 (2001); Fernandez et al, Cell Death Differ. 7:350 (2000)). Anti- apoptotic proteins such as Bcl-2 and/or BCI-XL are also thought to regulate cell-cell interactions, for example through regulation of cell surface integrins (Reed, Nature 387:773 (1997); Frisch et al, Curr. Opin. Cell Biol. 9:701 (1997); Del Bufalo et al. , FASEB J. 11:947 (1997)).
[0006] Therapeutic strategies for targeting Bcl-2 and BCI-XL in cancer to restore cancer cell sensitivity and overcome resistance of cancer cells to apoptosis have been extensively reviewed (Adams et al., Science 281:1322 (1998); Reed, Adv. Pharmacol. 41:501 (1997); Reed et al., J. Cell. Biochem. 60:23 (1996)).
[0007] Gossypol is a naturally occurring double biphenolic compound derived from crude cotton seed oil (Gossypium sp.). Human trials of gossypol as a male contraceptive have demonstrated the safety of long term administration of these compounds (Wu, Drugs 38:333 (1989)). Gossypol has more recently been shown to have some antiproliferative effects (Flack et al., J. Clin. Endocrinol. Metab. 76:1019 (1993); Bushunow et al., J. Neuro-Oncol. 43:79, (1999); Van Poznak et al, Breast Cancer Res. Treat. 66:239 (2001)). Gossypol and its derivatives recently have been shown to be potent inhibitors of Bcl-2 and BCI-XL and to have strong anti-cancer activity (U.S. Patent Application No. 2003/0008924).
SUMMARY OF THE INVENTION
[0008] The present invention relates to pulsatile dose administration of gossypol, i.e., (±)-gossypol (I), (-)-gossypol (II) or (+)-gossypol. It has surprisingly been found that pulsatile dose administration of gossypol provides clinical efficacy coupled with a reduction in adverse events. Gossypol and pharmaceutical compositions thereof are useful for treating hyperproliferative and other diseases, inhibiting the activity of anti-apoptotic Bcl-2 family proteins, inducing apoptosis in cells and increasing the sensitivity of cells to inducers of apoptosis.
Figure imgf000005_0001
Il
The present invention contemplates that pulsatile dose administration of gossypol to patients suffering from cancer and other diseases will expose patients to therapeutically effective amounts of gossypol and will minimize unwanted adverse events. Gossypol inhibits the function(s) of anti-apoptotic Bcl-2 family proteins and will kill cancer cells or supporting cells outright (those cells whose continued survival is dependent on the overactivity of Bcl-2 family proteins) and/or render such cells as a population more susceptible to the cell death-inducing activity of chemotherapeutic and/or radiotherapeutic agents. The present invention contemplates that gossypol administered by pulsatile dosing will satisfy an unmet need for the treatment of multiple cancer types, either when administered as monotherapy to induce apoptosis in cancer cells dependent on anti-apoptotic Bcl-2 family proteins function, or when administered in a temporal relationship with other cell death-inducing chemotherapeutic and/or radiotherapeutic agents so as to render a greater proportion of the cancer cells or supportive cells susceptible to executing the apoptosis program compared to the corresponding proportion of cells in a patient treated only with the chemotherapeutic and/or radiotherapeutic agent alone. The present invention further contemplates that gossypol administered by pulsatile dosing will induce apoptosis and/or render cells more sensitive to induction of apoptosis in other diseases or conditions characterized by dysregulation of apoptosis.
[0010] In certain embodiments of the invention, it is expected that combination treatment of patients with, a therapeutically effective amount of gossypol administered by pulsatile dosing and one or more additional therapeutic agents will produce a greater tumor response and clinical benefit in such patients compared to those treated with either gossypol or one or more therapeutic agents alone. Put another way, because gossypol administered by pulsatile dosing lowers the apoptotic threshold of all cells that express anti- apoptotic Bcl-2 family proteins, the proportion of cells that successfully execute the apoptosis program in response to the apoptosis inducing activity of therapeutic agents, such as anticancer drugs, will be increased. Alternatively, gossypol administered by pulsatile dosing is expected to allow administration of a lower, and therefore less toxic and more tolerable, dose of an anticancer agent to produce the same tumor response/clinical benefit as the conventional dose of the anticancer agent alone. Since the doses for all approved anticancer agents are known, the present invention contemplates combination therapies with various combinations of known anticancer agents with gossypol administered by pulsatile dosing. Also, since gossypol administered by pulsatile dosing acts at least in part by inhibiting anti-apoptotic Bcl-2 family proteins, the exposure of cancer cells and supporting cells to therapeutically effective amounts of gossypol can be temporally linked to coincide with the attempts of cells to execute the apoptosis program in response to the anticancer agent. Thus, in some embodiments, administering gossypol by pulsatile dosing in connection with certain temporal relationships, will provide especially efficacious therapeutic practices.
[0011] Gossypol administered by pulsatile dosing is useful for the treatment, amelioration, or prevention of disorders responsive to induction of apoptotic cell death, e.g., disorders characterized by dysregulation of apoptosis, including hyperproliferative diseases such as cancer. In certain embodiments, gossypol administered by pulsatile dosing can be used to treat, ameliorate, or prevent cancer that is characterized by resistance to cancer therapies (e.g., those which are chemoresistant, radiation resistant, hormone resistant, and the like). In one embodiment, the cancer is chemoresistant to treatment with a taxane, i.e., docetaxel or paclitaxel. In one embodiment, the chemoresistant cancer is prostate cancer. In additional embodiments, gossypol administered by pulsatile dosing can be used to treat, ameliorate, or prevent metastatic cancer. In other embodiments, gossypol administered by pulsatile dosing can be used to treat hyperproliferative and other diseases characterized by overexpression of anti-apoptotic Bcl-2 family proteins. In other embodiments, gossypol administered by pulsatile doing can be used to modulate spermicidal activity (e.g., function as a male contraceptive or antifertility agent), treat malaria, microbial or viral disease (e.g., inhibit the growth of the HIV virus as a treatment for AIDS), treat obesity, skin disorders or baldness, inhibit growth of endothelial cells, inhibit vascularization or neovascularization, treat arthritic conditions, neovascular-based dermatological conditions, diabetic retinopathy, Kaposi's sarcoma, age-related macular degeneration, restenosis, telangiectasia, glaucoma, keloids, corneal graft rejection, wound granularization, angiofibroma, Osier-Webber syndrome, myocardial angiogenesis, psoriatic arthritis or scleroderma, inhibit DNA synthesis or DNA polymerase activity and treat gynecological disorders (e.g., endometriosis) or diabetic complications.
BRIEF DESCRIPTION OF THE DRAWINGS/FIGURES
[0012] Fig. 1 is a chart showing pulsatile dose administration schedules of gossypol. [0013] Fig. 2 is a graph showing the in vivo activity of (-)-gossypol acetic acid co-crystal via pulse dose administration in combination with docetaxel. [0014] Fig. 3 is a graph showing the in vivo activity of (-)-gossypol acetic acid co-crystals via pulse dose administration in combination with docetaxel.
[0015] Fig, 4 is a waterfall plot showing PSA response in chemo-naive patients after treatment with (-)-gossypol acetic acid co-crystals.
[0016] Fig. 5 is a graph showing PSA response versus time in chemo-naive patients after treatment with (-)-gossypol acetic acid co-crystals.
[0017] Fig. 6 is a series of four images showing tumor size before and after treatment with (-)-gossypol acetic acid co-crystals.
[0018] Fig. 7 is a waterfall plot showing PSA response in docetaxel (taxotere)- resistant patients after treatment with (-)-gossypol acetic acid co-crystals.
[0019] Fig. 8 is a graph showing PSA response kinetics in a docetaxel
(taxotere)-resistant patient after treatment with (-)-gossypol acetic acid co- crystals.
[0020] Fig. 9 is a graph showing PSA response kinetics in a docetaxel
(taxotere)-resistant patient after treatment with (-)-gossypol acetic acid co- crystals.
[0021] Fig. 10 is a graph showing PSA response kinetics in a docetaxel
(taxotere)-resistant patient after treatment with (-)-gossypol acetic acid co- crystals.
DETAILED DESCRIPTION OF THE INVENTION
[0022] The present invention relates to pulsatile dose administration of gossypol. Various compositions comprising gossypol are contemplated for use in the methods of the invention. A gossypol composition may comprise, for example, (±)-gossypol, (-)-gossypol, (+)-gossypol, (±)-gossypol co- crystals, (-)-gossypol co-crystals or (+)-gossypol co-crystals. Gossypol is an inhibitor of anti-apoptotic Bcl-2 family proteins. By inhibiting anti-apoptotic Bcl-2 family proteins, gossypol sensitizes cells to inducers of apoptosis and, in some instances, itself induces apoptosis. Therefore, the invention relates to methods of sensitizing cells to inducers of apoptosis and to methods of inducing apoptosis in cells, comprising administering gossypol by pulsatile dosing alone or in combination with an additional therapeutic agent, such as an inducer of apoptosis. The invention further relates to methods of treating, ameliorating, or preventing disorders in a patient that are responsive to induction of apoptosis comprising administering to the patient gossypol by pulsatile dosing and an inducer of apoptosis, e.g., an anticancer agent, Such disorders include those characterized by a dysregulation of apoptosis and those characterized by overexpression of anti-apoptotic Bcl-2 family proteins. In one embodiment, the disease, condition or disorder responsive to the induction of apoptosis is selected from the group consisting of a hyperproliferative disease, i.e., cancer autoimmune disorder, chronic inflammatory condition, i.e., psoriasis, viral infection, microbial infection, parasitic infection, osteoarthritis, and atherosclerosis. In one embodiment, the cancer is selected from the group consisting of breast cancer, prostate cancer, lymphoma, skin cancer, pancreatic cancer, colon cancer, malignant melanoma, ovarian cancer, brain cancer, primary brain carcinoma, head-neck cancer, glioma, glioblastoma, liver cancer, bladder cancer, non-small cell lung cancer, head carcinoma, neck carcinoma, breast carcinoma, ovarian carcinoma, lung carcinoma, small-cell lung carcinoma, Wilms' tumor, cervical carcinoma, testicular carcinoma, bladder carcinoma, pancreatic carcinoma, stomach carcinoma, colon carcinoma, prostatic carcinoma, genitourinary carcinoma, thyroid carcinoma, esophageal carcinoma, myeloma, multiple myeloma, adrenal carcinoma, renal cell carcinoma, endometrial carcinoma, adrenal cortex carcinoma, malignant pancreatic insulinoma, malignant carcinoid carcinoma, choriocarcinoma, mycosis fungoides, malignant hypercalcemia, cervical hyperplasia, leukemia, acute lymphocytic leukemia, chronic lymphocytic leukemia, acute myelogenous leukemia, chronic myelogenous leukemia, chronic granulocytic leukemia, acute granulocytic leukemia, hairy cell leukemia, neuroblastoma, rhabdomyosarcoma, Kaposi's sarcoma, polycythemia vera, essential thrombocytosis, Hodgkin's disease, non-Hodgkin's lymphoma, soft-tissue sarcoma, osteogenic sarcoma, primary macroglobulinemia and retinoblastoma. In another embodiment, the cancer is prostate cancer.
[0024] The term "gossypol," as used herein refers to (±)-gossypol,
(-)-gossypol or (+)-gossypol, and pharmaceutically acceptable salts thereof, unless otherwise indicated.
[0025] The term "(-)-gossypol," as used herein, refers to an optically active composition of gossypol wherein the active molecules comprising the composition rotate plane polarized light counterclockwise (e.g., levorotatory molecules) as measured by a polarimeter. Preferably, the (-)-gossypol compound has an enantiomeric excess of 1% to 100%. In one embodiment, the (-)-gossypol compound has an enantiomeric excess of at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% (-)-gossypol. In one example of a "(-)-gossypol compound", the specific rotation ([α]o) of the compound is about -350° to about -390°, about -375° to about -390°, or about -385° to about -390°. (See e.g., Dowd, Chirality, 15:486 (2003); Ciesielska et al, Chem. Phys. Lett. 353:69 (2992); Freedman et al, Chirality, 15:196 (2003); and Zhou et al, Kexue Tongbao, 28:1574 (1983)). Methods for resolving racemic gossypol compounds into substantially purified (+)- or (-)-gossypol are known (See e.g., Zhou et al, Kexue Tongbao, 28:1574 (1983) (wherein: ^-phenylalanine methyl ester was mixed with the aldehyde groups of gossypol to form a Schiff s base with two diastereoisomers which were then resolved on a normal silica flash chromatography column. The filtrate was concentrated, and the residue was purified by chromatography on silica gel eluting with hexanes:EtOAc=3:l to give two fractions. Acid hydrolysis of the two fractions in 5N HC1:THF (1 :5, room temperature, overnight) regenerated the individual gossypol enantiomers, respectively. The first fraction with a higher Rf value contained (-)-gossypol, and the second fraction with a lower Rf value contained (+)-gossypol. The crude gossypol fractions were extracted into ether from the residue after removing THF from the reaction mixture. The gossypol fractions were then purified by chromatography on silica gel and eluted with hexanes:EtOAc (3:1 ratio) to give optically pure gossypol, with a yield of 30-40% in two steps. The optical rotatory dispersion values for these products were αD= -352° (c=0.65, CHCl3) for (-)-gossypol, and αD= +341° (c= 0.53, CHCl3)).
[0026] The term "C1-8 carboxylic acid," as used herein, refers to straight- chained or branched, aromatic or non-aromatic, saturated or unsaturated, substituted or unsubstituted C1-8 carboxylic acid, including, but not limited to, formic acid, acetic acid, propionic acid, n-butyric acid, t-butyric acid, n- pentanoic acid, 2-pentanoic acid, n-hexanoic acid, 2-hexanoic acid, n- heptanoic acid, n-octanoic acid, acrylic acid, succinic acid, fumaric acid, malic acid, tartaric acid, citric acid, lactic acid, and benzoic acid.
[0027] The term "Ci-8 sulfonic acid," as used herein, refers to straight-chained or branched, aromatic or non-aromatic, saturated or unsaturated, substituted or unsubstituted C1-8 sulfonic acid, including, but not limited to, methanesulfonic acid, ethanesulfonic acid, n-propanesulfonic acid, 2-propanesulfonic acid, n- butanesulfonic acid, n-pentanesulfonic acid n-hexanesulfonic acid, n- heptanesulfonic acid, n-octanesulfonic acid, and benzenesulfonic acid.
[0028] The term "C1-I2 ketone," as used herein, refers to straight-chained, cyclic or branched, aromatic or non-aromatic, saturated or unsaturated, substituted or unsubstituted Ci-12 ketone, including, but not limited to, acetone and cyclododecanone.
[0029] The term "Ci-12 diketone," as used herein, refers to straight-chained or branched, aromatic or non-aromatic, saturated or unsaturated, substituted or unsubstituted C1-12 diketone, including, but not limited to, 2,4-pentanedione.
[0030] The term "gossypol co-crystal," as used herein, refers to a composition comprising co-crystals of (±)-gossypol and a Ci-8 carboxylic acid, C1-8 sulfonic acid, CM2 ket one or CM2 diketone. The term "(-)-gossypol co- crystal," as used herein, refers to a composition comprising (-)-gossypol and acetic acid, (-)-gossypol and acetone, (-)-gossypol and 2,4-pentanedione or (-)- gossypol and cyclododecanone. In one embodiment, the (-)-gossypol co- crystal is (-)-gossypol acetic acid co-crystal. See U.S. 7,342,046. A method of preparing (-)-gossypol acetic acid co-crystal is described in U.S. 2008/0021110.
[0031] The term "pulsatile dose administration," as used herein, refers to intermittent (i.e., not constant daily) administration of gossypol. Pulsatile dose administration schedules useful in the present invention encompass any discontinuous daily administration regimen that provides a therapeutically effective amount of gossypol to a patient in need thereof. Pulsatile dosing regimens may use equivalent, lower or higher doses of gossypol than typically used in continuous daily dosing regimens. On the days that gossypol is scheduled to be administered, administration of gossypol may occur once a day, twice-a-day (i.e., BID), three times a day, four times a day or more in accordance with an intermittent daily dosing schedule.
[0032] The therapeutic utility of drug administration can be offset by the number and severity of adverse events a patient experiences. Pulsatile dosing of gossypol results in the unexpected combination of a reduction in the number and/or severity of clinical adverse events coupled with a maintenance or enhancement in clinical efficacy, as compared to continuous daily dosing. Moreover, the surprising clinical benefits of pulsatile dose administration of gossypol may be even more prominent when combined with other therapeutic agents.
[0033] In one example, gossypol may be administered no more frequently than one day out of every two days (i.e., day 1, day 3, day 5, day 7, day 9, etc.), every three days (i.e., day 1, day 4, day 7, day 10, etc.), every four days, every five days, every six days, every seven days, every eight days, every nine days, every ten days, every two weeks, every three weeks, every four weeks, or longer. The administration of gossypol can continue for one, two, three or four weeks, one, two, three or four months, one, two, three or four years or longer.
[0034] In another example, gossypol may be administered on a least two consecutive days, e.g., at least three, four, five, six or seven consecutive days, followed by at least one day, at least two consecutive days, at least three consecutive days, at least four consecutive days, at least five consecutive days, at least six consecutive days, at least seven consecutive days, at least eight consecutive days, at least nine consecutive days, at least ten consecutive days, at least eleven consecutive days, at least twelve consecutive days, at least thirteen consecutive days, at least two consecutive weeks, at least three consecutive weeks or at least four consecutive weeks or longer wherein gossypol is not administered. The administration of gossypol can continue for one, two, three or four weeks, one, two, three or four months, one, two, three or four years or longer.
[0035] In another example, gossypol may be administered twice-a-day for three consecutive days followed by eleven, twelve, thirteen, fourteen, fifteen, sixteen, seventeen, eighteen, nineteen or twenty consecutive days, or longer, wherein gossypol is not administered. In a particular example, a pharmaceutical composition comprising (-)-gossypol may be administered twice-a-day for three consecutive days followed by seventeen or eighteen consecutive days wherein gossypol is not administered. The administration of gossypol can continue for two, three or four weeks, one, two, three or four months, one, two, three or four years or longer.
[0036] In another example, gossypol may be administered one day a week, e.g., gossypol administered on one day followed by six consecutive days wherein gossypol is not administered, for at least two weeks, at least three weeks, at least four weeks, at least five weeks, at least six weeks, at least seven weeks or at least eight weeks. The administration of gossypol can continue for one, two, three or four months, one, two, three or four years or longer.
[0037] In another example, gossypol may be administered via the sequential use of a combination of two or more pulsatile dosing schedules. The combination may comprise the same pulsatile dosing schedules, e.g., gossypol administered twice-a-day for three consecutive days followed by eleven consecutive days wherein gossypol is not administered followed by gossypol administered twice-a-day for three consecutive days followed by eleven consecutive days wherein gossypol is not administered, or different pulsatile dosing schedules, e.g., gossypol administered twice-a-day for three consecutive days followed by eleven consecutive days wherein gossypol is not administered followed by gossypol administered on one day followed by six consecutive days wherein gossypol is not administered. In one example, pulsatile dose administration of gossypol comprises administration on one day followed by six consecutive days wherein gossypol is not administered followed by administration on at least two consecutive days followed by at least one day wherein gossypol is not administered. In another example, pulsatile dose administration of gossypol comprises administration twice-a- day for at least three consecutive days followed by at least seven consecutive days wherein gossypol is not administered followed by administration on one day followed by at least six consecutive days wherein gossypol is not administered. In another example, pulsatile dose administration of gossypol comprises administration on one day followed by six consecutive days wherein gossypol is not administered followed by administration on one day followed by one day wherein gossypol is not administered. The sequential use of a combination of two or more pulsatile dosing regimens may be repeated as many times as necessary to achieve or maintain a therapeutic response, e.g., from one to about fifty times, e.g., from one to about twenty times, e.g., from about one to about ten times. With every repetition any additional therapeutic agents may be the same or different from that used in the previous repetition. In another embodiment of the invention, gossypol may be administered according to a pulsatile dosing schedule and/or sequential combination of two or more pulsatile dosing schedules followed by a waiting period. The term "waiting period," as used herein, refers to a period of time between dosing schedules when gossypol is not administered to the patient. The waiting period may be one, two, three, four, five or six days, one, two or three weeks, one, two, three or four months, one, two, three or four years or longer. In certain embodiments, the waiting period may be one to thirty days, e.g., seven, fourteen, twenty one or thirty days, e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29 or 30 days. After the waiting period, the same or a different pulsatile dosing schedule and/or sequential combination of one or more pulsatile dosing schedules of gossypol may resume. In one example, pulsatile dose administration of gossypol comprises administration on one day followed by six consecutive days wherein gossypol is not administered, a waiting period and administration on at least two consecutive days followed by at least one day wherein gossypol is not administered. In another example, pulsatile administration of gossypol comprises administration on one day followed by six consecutive days wherein gossypol is not administered, a waiting period and administration on one day followed by six consecutive days wherein gossypol is not administered. In another example, pulsatile dose administration of gossypol comprises administration twice-a-day for at least three consecutive days followed by at least seven consecutive days wherein gossypol is not administered, a waiting period and administration on one day followed by at least six consecutive days wherein gossypol is not administered. The pulsatile dosing/waiting period regimen may be repeated as many times as necessary to achieve or maintain a therapeutic response, e.g., from one to about fifty times, e.g., from one to about twenty times, e.g., from about one to about ten times. With every repetition any additional therapeutic agents may be the same or different from that used in the previous repetition.
[0039] Illustrative pulsatile dose administration schedules are shown in Figure
1. In addition, referring to Figure 1, gossypol may be administered, for example, via the sequential use of a combination of schedule A and B, schedule B and C, schedule C and D, schedule D and E, schedule F and G, schedule G and H, schedule H and I, schedule H and H, schedule I and I, schedule H, I and H or schedule G, H and I. Also, gossypol may be administered via schedule H followed by a waiting period {e.g., one to thirty days) followed by administration via schedule I, schedule A-waiting period- schedule B, schedule H- waiting period-schedule E- waiting period-schedule A, schedule H-waiting period-schedule I-waiting period-schedule H or schedule H-schedule I-waiting period-schedule H-schedule I. The above-described pulsatile dose administration schedules are provided for illustrative purposes only and should not be considered limiting. The present invention contemplates any discontinuous daily administration regimen that provides a therapeutically effective amount of gossypol to a patient in need thereof.
[0040] The term "adverse event," as used herein, refers to any undesirable change in health that occurs to a patient during a clinical trial or within a period of time after the clinical trial is complete. Adverse events are categorized by grade, with less serious adverse events given grades 1 (mild) and 2 (moderate) and more serious adverse events given grades 3 (severe) and 4 (life-threatening or disabling). The grading of adverse events can be done using any scale known in the art, such as the National Cancer Institute scale (Common Terminology Criteria for Adverse Events, v3.0). A decrease in the number of adverse events refers to a decrease in the actual number of events. A decrease in the severity of adverse events refers to a decrease in the grade of the adverse events that occur.
[0041] The term "Bcl-2 family proteins," as used herein, refers to both the anti-apoptotic members of the Bcl-2 family, including, but not limited to, BcI- 2, BcI-XL, McI-I, Al/BFL-1, BOO-DIVA, Bcl-w, Bcl-6, Bcl-8, and Bcl-y, and the pro-apoptotic members of the Bcl-2 family, including, but not limited to, Bak, Bax, Bad, tBid, Hrk, Bim, Bmf, as well as other Bcl-2 homology domain 3 (BH3) containing proteins that are regulated by gossypol compounds.
[0042] The term "overexpression of anti-apoptotic Bcl-2 family proteins," as used herein, refers to an elevated level (e.g., aberrant level) of mRNAs encoding for an anti-apoptotic Bcl-2 family protein(s), and/or to elevated levels of anti-apoptotic Bcl-2 family protein(s) in cells as compared to similar corresponding non-pathological cells expressing basal levels of mRNAs encoding anti-apoptotic Bcl-2 family proteins or having basal levels of anti- apoptotic Bcl-2 family proteins. Methods for detecting the levels of mRNAs encoding anti-apoptotic Bcl-2 family proteins or levels of anti-apoptotic Bcl-2 family proteins in a cell include, but are not limited to, Western blotting using anti-apoptotic Bcl-2 family protein antibodies, immunohistochemical methods, and methods of nucleic acid amplification or direct RNA detection. As important as the absolute level of anti-apoptotic Bcl-2 family proteins in cells is to determining that they overexpress anti-apoptotic Bcl-2 family proteins, so also is the relative level of anti-apoptotic Bcl-2 family proteins to other pro- apoptotic signaling molecules (e.g., pro-apoptotic Bcl-2 family proteins) within such cells. When the balance of these two are such that, were it not for the levels of the anti-apoptotic Bcl-2 family proteins, the pro-apoptotic signaling molecules would be sufficient to cause the cells to execute the apoptosis program and die, said cells would be dependent on the anti-apoptotic Bcl-2 family proteins for their survival. In such cells, exposure to an inhibiting effective amount of an anti-apoptotic Bcl-2 family protein inhibitor will be sufficient to cause the cells to execute the apoptosis program and die. Thus, the term "overexpression of an anti-apoptotic Bcl-2 family protein" also refers to cells that, due to the relative levels of pro-apoptotic signals and anti- apoptotic signals, undergo apoptosis in response to inhibiting effective amounts of compounds that inhibit the function of anti-apoptotic Bcl-2 family proteins.
[0043] The term "therapeutic agent," as used herein, refers to any agent which can be used in the treatment, management, or amelioration of a disease, condition or disorder or one or more symptoms thereof. In certain embodiments, the term "therapeutic agent" refers to gossypol, e.g., (-)-gossypol. In certain other embodiments, the term "therapeutic agent" does not refer to gossypol. Preferably, a therapeutic agent is an agent which is known to be useful for, or has been or is currently being used for the treatment, management, prevention, or amelioration of a disorder or one or more symptoms thereof. In one embodiment, the therapeutic agent is an anticancer agent.
[0044] The terms "anticancer agent" and "anticancer drug" as used herein, refer to any therapeutic agent (e.g., chemotherapeutic compounds and/or molecular therapeutic compounds), radiation therapies, or surgical interventions, used in the treatment of hyperproliferative diseases such as cancer (e.g., in mammals).
[0045] The term "therapeutically effective amount," as used herein, refers to that amount of the therapeutic agent sufficient to result in amelioration of one or more symptoms of a disorder, or prevent advancement of a disorder, or cause regression of the disorder. For example, with respect to the treatment of cancer, a therapeutically effective amount preferably refers to the amount of a therapeutic agent that decreases the rate of tumor growth, decreases tumor mass, decreases the number of metastases, increases time to tumor progression, or increases survival time by at least 5%, preferably at least 10%, at least 15%, at least 20%, at least 25%, at least 30%, at least 35%, at least 40%, at least 45%, at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, or at least 100%.
[0046] The terms "sensitize" and "sensitizing," as used herein, refer to making, through the administration of gossypol, a patient or a cell within a patient more susceptible, or more responsive, to the biological effects (e.g., promotion or retardation of an aspect of cellular function including, but not limited to, cell growth, proliferation, invasion, angiogenesis, or apoptosis) of a second therapeutic agent. The sensitizing effect of gossypol on a target cell can be measured as the difference in the intended biological effect (e.g., promotion or retardation of an aspect of cellular function including, but not limited to, cell growth, proliferation, invasion, angiogenesis, or apoptosis) observed upon the administration of a second therapeutic agent with and without administration of gossypol. The response of the sensitized cell can be increased by at least 5 %, at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 100%, at least 150%, at least 200%, at least 350%, at least 300%, at least 350%, at least 400%, at least 450%, or at least 500% over the response in the absence of gossypol. [0047] The term "dysregulation of apoptosis," as used herein, refers to any aberration in the ability of (e.g., predisposition) a cell to undergo cell death via apoptosis. Dysregulation of apoptosis is associated with or induced by a variety of conditions, including for example, autoimmune disorders (e.g., systemic lupus erythematosus, rheumatoid arthritis, graft-versus-host disease, myasthenia gravis, or Sjogren's syndrome), chronic inflammatory conditions (e.g., psoriasis, asthma or Crohn's disease), hyperproliferative disorders (e.g., tumors, B cell lymphomas, or T cell lymphomas), viral infections (e.g., herpes, papilloma, or HIV), microbial infections, parasitic infections and other conditions such as osteoarthritis and atherosclerosis. It should be noted that when the dysregulation is induced by or associated with a viral infection, the viral infection may or may not be detectable at the time dysregulation occurs or is observed. That is, viral-induced dysregulation can occur even after the disappearance of symptoms of viral infection.
[0048] The term "hyperproliferative disease," as used herein, refers to any condition in which a localized population of proliferating cells in an animal is not governed by the usual limitations of normal growth, such as cancer. Examples of hyperproliferative disorders include psoriasis, restenosis, tumors, neoplasms, lymphomas and the like. A neoplasm is said to be benign if it does not undergo invasion or metastasis and malignant if it does either of these. A "metastatic" cell means that the cell can invade and destroy neighboring body structures. Hyperplasia is a form of cell proliferation involving an increase in cell number in a tissue or organ without significant alteration in structure or function. Metaplasia is a form of controlled cell growth in which one type of fully differentiated cell substitutes for another type of differentiated cell.
[0049] The term "cancer," as used herein, is intended to refer to any known cancer, and may include, but is not limited to the following: leukemias such as acute leukemia, acute lymphocytic leukemia, acute myelocytic leukemias such as myeloblastic, promyelocytic, myelomonocytic, monocytic, and erythroleukemia leukemias, and myelodysplastic syndrome; chronic leukemias such as chronic myelocytic (granulocytic) leukemia, chronic lymphocytic leukemia, and hairy cell leukemia; polycythemia vera; lymphomas such as Hodgkin's disease and non-Hodgkin's disease; multiple myelomas such as smoldering multiple myeloma, non-secretory myeloma, osteosclerotic myeloma, plasma cell leukemia, solitary plasmacytoma and extramedullary plasmacytoma; Waldenstrom's macroglobulinemia; monoclonal gammopathy of undetermined significance; benign monoclonal gammopathy; heavy chain disease; bone and connective tissue sarcomas such as bone sarcoma, osteosarcoma, chondrosarcoma, Ewing's sarcoma, malignant giant cell tumor, fibrosarcoma of bone, chordoma, periosteal sarcoma, soft-tissue sarcomas, angiosarcoma (hemangiosarcoma), fibrosarcoma, Kaposi's sarcoma, leiomyosarcoma, liposarcoma, lymphangiosarcoma, neurilemmoma, rhabdomyosarcoma, and synovial sarcoma; brain tumors such as glioma, astrocytoma, brain stem glioma, ependymoma, oligodendroglioma, nonglial tumor, acoustic neurinoma, craniopharyngioma, medulloblastoma, meningioma, pineocytoma, pineoblastoma, and primary brain lymphoma; breast cancers such as adenocarcinoma, lobular (small cell) carcinoma, intraductal carcinoma, medullary breast cancer, mucinous breast cancer, tubular breast cancer, papillary breast cancer, Paget's disease of the breast, and inflammatory breast cancer; adrenal cancers such as pheochromocytoma and adrenocortical carcinoma; thyroid cancers such as papillary or follicular thyroid cancer, medullary thyroid cancer and anaplastic thyroid cancer; pancreatic cancers such as insulinoma, gastrinoma, glucagonoma, vipoma, somatostatin-secreting tumor, and carcinoid or islet cell tumor; pituitary cancers such as prolactin-secreting tumor and acromegaly; eye cancers such as ocular melanoma, iris melanoma, choroidal melanoma, and cilliary body melanoma, and retinoblastoma; vaginal cancers such as squamous cell carcinoma, adenocarcinoma, and melanoma; vulvar cancers such as squamous cell carcinoma, melanoma, adenocarcinoma, basal cell carcinoma, sarcoma, and Paget's disease of the genitals; cervical cancers such as squamous cell carcinoma and adenocarcinoma; uterine cancers such as endometrial carcinoma and uterine sarcoma; ovarian cancers such as ovarian epithelial carcinoma, ovarian epithelial borderline tumor, germ cell tumor, and stromal tumor; esophageal cancers such as squamous cancer, adenocarcinoma, adenoid cyctic carcinoma, mucoepidermoid carcinoma, adenosquamous carcinoma, sarcoma, melanoma, plasmacytoma, verrucous carcinoma, and oat cell (small cell) carcinoma; stomach cancers such as adenocarcinoma, fungating (polypoid), ulcerating, superficial spreading, diffusely spreading, malignant lymphoma, liposarcoma, fibrosarcoma, and carcinosarcoma; colon cancers; rectal cancers; liver cancers such as hepatocellular carcinoma and hepatoblastoma, gallbladder cancers such as adenocarcinoma; cholangiocarcinomas such as papillary, nodular, and diffuse; lung cancers such as non-small cell lung cancer, squamous cell carcinoma (epidermoid carcinoma), adenocarcinoma, large-cell carcinoma and small-cell lung cancer; testicular cancers such as germinal tumor, seminoma, anaplastic, classic (typical), spermatocyte, nonseminoma, embryonal carcinoma, teratoma carcinoma, and choriocarcinoma (yolk-sac tumor), prostate cancers such as adenocarcinoma, leiomyosarcoma, and rhabdomyosarcoma; penile cancers; oral cancers such as squamous cell carcinoma; basal cancers; salivary gland cancers such as adenocarcinoma, mucoepidermoid carcinoma, and adenoidcystic carcinoma; pharynx cancers such as squamous cell cancer and verrucous; skin cancers such as basal cell carcinoma, squamous cell carcinoma and melanoma, superficial spreading melanoma, nodular melanoma, lentigo malignant melanoma, acral lentiginous melanoma; head and neck cancers; kidney cancers such as renal cell cancer, adenocarcinoma, hypernephroma, fibrosarcoma, transitional cell cancer (renal pelvis and/or ureter); Wilms' tumor; and bladder cancers such as transitional cell carcinoma, squamous cell cancer, adenocarcinoma, and carcinosarcoma. In addition, cancers that can be treated by the methods and compositions of the present invention include myxosarcoma, osteogenic sarcoma, endotheliosarcoma, lymphangioendotheliosarcoma, mesothelioma, synovioma, hemangioblastoma, epithelial carcinoma, cystadenocarcinoma, bronchogenic carcinoma, sweat gland carcinoma, sebaceous gland carcinoma, papillary carcinoma and papillary adenocarcinoma. See Fishman et ah, 1985, Medicine, 2d Ed., J.B. Lippincott Co., Philadelphia, PA and Murphy et ah, 1997, Informed Decisions: The Complete Book of Cancer Diagnosis, Treatment, and Recovery, Viking Penguin, New York, NY, for a review of such disorders.
[0050] The pathological growth of activated lymphoid cells often results in an autoimmune disorder or a chronic inflammatory condition. As used herein, the term "autoimmune disorder" refers to any condition in which an organism produces antibodies or immune cells which recognize the organism's own molecules, cells or tissues. Non-limiting examples of autoimmune disorders include autoimmune hemolytic anemia, autoimmune hepatitis, Berger's disease or IgA nephropathy, celiac sprue, chronic fatigue syndrome, Crohn's disease, dermatomyositis, fibromyalgia, graft versus host disease, Grave's disease, Hashimoto's thyroiditis, idiopathic thrombocytopenia purpura, lichen planus, multiple sclerosis, myasthenia gravis, psoriasis, rheumatic fever, rheumatic arthritis, scleroderma, Sjogren's syndrome, systemic lupus erythematosus, type 1 diabetes, ulcerative colitis, vitiligo, and the like.
[0051] The term "neoplastic disease," as used herein, refers to any abnormal growth of cells being either benign (non-cancerous) or malignant (cancerous).
[0052] The term "anti-neoplastic agent," as used herein, refers to any compound that retards the proliferation, growth, or spread of a targeted {e.g., malignant) neoplasm.
[0053] The terms "prevent," "preventing" and "prevention," as used herein, refer to a decrease in the occurrence of pathological cells {e.g., hyperproliferative or neoplastic cells) in an animal. The prevention may be complete, e.g., the total absence of pathological cells in a subject. The prevention may also be partial, such that the occurrence of pathological cells in a subject is less than that which would have occurred without the present invention.
[0054] The term "synergistic," as used herein, refers to an effect obtained when gossypol and a second therapeutic agent are administered together {e.g., at the same time or one after the other) that is greater than the additive effect of gossypol and the second therapeutic agent when administered individually. The synergistic effect allows for lower doses of gossypol and/or the second therapeutic agent to be administered or provides greater efficacy at the same doses. The synergistic effect obtained can be at least 5 %, at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 100%, at least 125%, at least 150%, at least 175%, at least 200%, at least 250%, at least 300%, at least 350%, at least 400%, or at least 500% more than the additive effect of gossypol and the second therapeutic agent when administered individually. For example, with respect to the treatment of cancer, the synergistic effect can be a decrease in the rate of tumor growth, a decrease in tumor mass, a decrease in the number of metastases, an increase in time to tumor progression, or an increase in survival time. The co-administration of gossypol by pulsatile dosing and an anticancer agent may allow for the use of lower doses of gossypol and/or the anticancer agent such that the cancer is effectively treated while avoiding any substantial toxicity to the subject.
[0055] The term "about," as used herein, includes the recited number +/- 10%.
Thus, "about 0.5" means 0.45 to 0.55.
[0056] Inhibitors of anti-apoptotic Bcl-2 family proteins useful in the present invention include, for example, gossypol or co-crystals of gossypol or pharmaceutical compositions thereof that are administered by pulsatile dosing. Those skilled in the art will appreciate the importance of compound stability in the manufacturing, storage, shipping, and/or handling of pharmaceutical compositions. Compositions comprising co-crystals of gossypol may be more stable than some other compositions comprising gossypol. Any Ci-8 carboxylic acid or Cj-8 sulfonic acid that is capable of stabilizing gossypol may be used in the composition. The molar ratio of gossypol to carboxylic acid or sulfonic acid in gossypol co-crystal ranges from about 10:1 to about 1 :10, e.g., about 2:1 to about 1 :2, e.g., about 1 :1. [0057] Compositions comprising gossypol useful in this invention may be prepared using methods known to those of skill in the art, such as the methods disclosed in U.S. Published Application No. 2005/0234135. For example, (-)- gossypol co-crystal may be prepared by dissolving (-)-gossypol in acetone to form a solution, filtering the solution, adding acetic acid into the solution with mixing until the solution turns turbid, leaving the turbid solution at room temperature and then at reduced temperature to form co-crystals, collecting the co-crystals, washing the co-crystals with a solvent, and drying the co-crystals. Reduced temperature is less than about 2O0C, preferably about 0-150C, more preferably about 4°C. The time for co-crystal formation may range from 1 hour to 1 day; preferably the time is about 1-4 hours. The co-crystals may be collected by any suitable means, including by filtration. The solvent for washing the co-crystals may be any suitable solvent, e.g., hexane, pentane, benzene, toluene, or petroleum ether. The washed co-crystals may be dried at room temperature, preferably in a lightproof container. The co-crystals may also be dried in a vacuum drier, preferably at an elevated temperature {e.g., about 30-600C, more preferably about 4O0C) for about 6-72 hours, preferably about 12-48 hours.
[0058] Gossypol acetic acid co-crystals may also be prepared from a mixture of (±)-gossypol and acetic acid via recrystallization. The gossypol acetic acid co-crystals may be further recrystallized from a solution of gossypol acetic acid in a mixture of acetone and acetic acid. The recrystallization mixture is held for about 15 minutes to about 100 minutes, e.g., about 30 minutes to about 60 minutes, to allow co-crystal formation. The recrystallization is carried out at ambient temperature, e.g., about 150C to about 3O0C, e.g., about 220C. Following the recrystallization, the crystals are harvested from the recrystallization mixture (e.g., by filtration) and washed with a non-polar solvent, e.g., pentane, hexene, hexane(s), heptane, or mixtures thereof. Preferably, the washing step is quick to avoid the incorporation of the non- polar solvent into the crystals. Short washing times (less than 2 minutes) are preferred. The crystals may then be dried, e.g., in vacuo, while protected from light. The recrystallization may be repeated more than once (e.g., 2, 3, 4, 5, or more times) to improve the impurity profile, e.g., until gossypol co-crystals comprise less than about 5%, 4%, 3%, 2%, 1%, 0.5%, or 0.1% impurities. Once the desired impurity profile is obtained, a final recrystallization may be carried out at a lower temperature, e.g., about -2O0C to about 1O0C, preferably about -1O0C to about 00C. The recrystallization mixture is held for about 15 minutes to about 100 minutes, e.g., about 30 to about 60 minutes, and the resulting crystals are then filtered, washed and dried as described above. This method of producing gossypol acetic acid co-crystals produces a final product comprising less than about 5% total impurities, preferably less than about 3%, 2%, or 1%. (-)-Gossypol acetic acid co-crystals may also be prepared from gossypol acetic acid co crystals via imine (Schiff base) formation with an optically active amine to form diastereomers. As used herein, the term "imine" includes other tautomers such as eneamine tautomers and stereoisomers thereof. The optically active amine may be, but is not limited to, L-phenylalanine methyl ester, S-1-methylphenethylamine, or L- phenylalaninol or the corresponding HCl salt. The derivatization may be carried out in the absence of oxygen, e.g., under a nitrogen purge. The derivatization is carried out in the presence of a nonpolar and/or polar solvent, e.g., dichloromethane and/or isopropanol, for a time period of about 0.5 to about 3 hours, e.g., about 1 hour to about 2 hours. A dehydrating agent such as sodium sulfate or a molecular sieve, e.g., type 3 A, is then added, along with suitable reagents for buffering the reaction mix at a pH of about 5 to about 7, e.g., about 6. One suitable buffering agent is sodium bicarbonate. The reaction mixture is then stirred for at least about 15 minutes, e.g., at least about 30 minutes. The progression of the reaction may be monitored for completion. For example, the reaction mixture may be assayed for the absence of gossypol using thin layer chromatography (TLC) or preferably in real time by high pressure liquid chromatography (HPLC). If the reaction is incomplete, the pH may be adjusted back to about 6 by adding further buffering agents. The reaction is then continued for about 24 hours and again checked for completion of the reaction. After completion, the reaction mixture may be filtered to remove the solids and the solids washed with additional non-polar solvent, e.g., dichloromethane. The filtrates may then be evaporated to dryness, e.g., with a rotary evaporator with the bath set at about 3O0C to about 4O0C.
[0060] The resultant diastereomers are then separated, e.g., by chromatography For example, the diastereomers are separated by silica gel chromatography, e.g., Kromasil Si. The dried filtrate is reconstituted in a non- polar solvent, e.g., dichloromethane, and charged onto the column. The diastereomers are eluted with a solvent system comprising non-polar and polar solvents, e.g., 1 :1 heptane:ethyl acetate. Column fractions may be monitored by HPLC and fractions containing the desired isomer (e.g., at least about 90%) may be pooled and evaporated. Impure fractions may be collected and passed over the column additional times. In one embodiment, the diastereomer mixture is held for less than 24 hours, preferably less than 12, 6, or 3 hours, prior to separation in order to avoid any increase in contamination with gossypol derivatives.
[0061] The separated R-(-)-gossypol bis-imine diastereomer may then be hydrolyzed to produce (-)-gossypol. The hydrolysis may be carried out in the absence of oxygen, e.g., under a nitrogen purge. The (-)-gossypol derivative is mixed with a polar solvent (e.g., tetrahydrofuran) and an acid (e.g., aqueous hydrochloric acid) and stirred for at least about 1 hour, e.g., at least about 5 hours. The extent of the reaction may be monitored by TLC or HPLC for depletion of both the bis- and mono-imine compounds to less than about 10%, preferably less than about 5%. If the reaction is not sufficient, it may be continued for at least about 15 hours and re-evaluated.
[0062] Once sufficient hydrolysis has occurred, the reaction mixture may then be washed with an aqueous brine solution. The aqueous brine solution may be back extracted with a polar solvent (e.g., ethyl acetate). The organic layers are then combined and washed with an alkaline aqueous solution (e.g., sodium bicarbonate) followed by a brine solution. The organic layers may then be evaporated to dryness, e.g., with a rotary evaporator with the bath set at about 3O0C to about 400C.
[0063] The crude isolate is then dissolved in a solvent system comprising non- polar and polar solvents {e.g., 1 :1 heptane: ethyl acetate) and passed over a silica gel plug using the same solvent system containing a small amount of acetic acid (to avoid sticking to the plug). Fractions may be collected and monitored for gossypol content using TLC or HPLC. Product-containing fractions may be pooled and evaporated to dryness, e.g., with a rotary evaporator with the bath set at about 3O0C to about 4O0C.
[0064] If further purification of the (-)-gossypol is desired, the (-)-gossypol may be purified by chromatography over a hydrophilic resin, e.g., a dihydroxypropyl resin such as DIOL, e.g., YMC DIOL (120 angstrom x 10-20 micron) (GL Sciences). The dried product from the previous step may be reconstituted in a solvent system comprising non-polar and polar solvents (e.g., 1 :1 heptane:ethyl acetate) and purified over the column using the same solvent system. Fractions are collected, held at a reduced temperature (e.g., about 2°C to about 8°C), and the fractions assayed for gossypol content using TLC or HPLC. Fractions containing gossypol (e.g., at least 90%) may be pooled and evaporated to dryness, e.g., with a rotary evaporator with the bath set at about 3O0C to about 4O0C. Fractions with less than 90% gossypol may be pooled and re-purified over the column.
[0065] As a final step, the purified (-)-gossypol may be dissolved in acetone
(e.g., at about 4 mL per 1 g gossypol) and glacial acetic acid is added (about 1.5 mL per 1 g gossypol). The mixture may then be loaded into a suitable container for crystallization (e.g., a Bϋchi Ball). If there is no immediate crystallization, the solvent may be slowly removed by vacuum until a crystal mass appears. The mixture may then be held for about 15 minutes to about 100 minutes, e.g., about 30 minutes to about 60 minutes, and then filtered. The crystals may then be washed with the same ratio of acetone and acetic acid. Finally, the crystals may be soaked in acetic acid (about 3 mL per 1 g gossypol for about 20 to about 40 minutes, preferably about 30 minutes, and the acetic acid removed by filtration. The crystals may then be dried (e.g. , in vacuo) for at least one hour, e.g., about 2 to about 4 hours. The crystals may be packaged and stored protected from light (e.g., in amber glass vials) at a reduced temperature (e.g., about -3O0C to about 00C, preferably about -100C to about -2O0C.
[0066] An alternative method of derivatizing gossypol acetic acid starting material comprising (±)-gossypol to form a Schiff base in the above-described methods of producing (-)-gossypol acetic acid co-crystals comprises treating a mixture of gossypol acetic acid starting material and optically active amine (e.g., L-phenylalanine methyl ester hydrochloride) in a nonpolar solvent (e.g., dichloromethane) with triethylamine and mixing for at least 2 hours, e.g., about 5 hours, optionally under an oxygen-free atmosphere. The reaction may be monitored for completion using HPLC or TLC methods. After completion of the reaction, the mixture is extracted with water and the organic phase separated and evaporated to dryness, e.g., with a rotary evaporator with the bath set at about 25°C to about 35°C, followed by a high vacuum overnight.
[0067] Gossypol been shown to bind to Bcl-2 and BCI-XL at the BH3 binding groove and to have anticancer activity (U.S. Patent Application No. 2003/0008924). Thus, gossypol may be used to induce apoptosis and also potentiate the induction of apoptosis in response to apoptosis induction signals when administered by pulsatile dosing. It is contemplated that gossypol administered by pulsatile dosing will sensitize cells to inducers of apoptosis, including cells that are resistant to such inducers. Gossypol administered by pulsatile dosing can be used to induce apoptosis in any disorder that can be treated, ameliorated, or prevented by the induction of apoptosis. Thus, the present invention provides methods for targeting patients characterized as overexpressing an anti-apoptotic Bcl-2 family protein. In some of the embodiments, the cells (e.g., cancer cells) show elevated expression levels of one or more anti-apoptotic Bcl-2 family proteins as compared to non- pathological samples (e.g., non-cancerous cells). In other embodiments, the cells operationally manifest elevated expression levels of anti-apoptotic Bcl-2 family proteins by virtue of executing the apoptosis program and dying in response to administration of an inhibiting effective amount of gossypol, said response occurring, at least in part, due to the dependence in such cells on anti-apoptotic Bcl-2 family protein function for their survival.
[0068] In some embodiments, the methods of the present invention are used to treat diseased cells, tissues, organs, or pathological conditions and/or disease states in a patient (e.g., a mammalian subject including, but not limited to, humans and veterinary animals). In this regard, various diseases and pathologies are amenable to treatment or prophylaxis using the present methods. A non-limiting exemplary list of these diseases and conditions includes, but is not limited to, cancers, T and B cell mediated autoimmune diseases, inflammatory diseases, infections, hyperproliferative diseases, AIDS, degenerative conditions, vascular diseases, and the like. In some embodiments, the cancer cells being treated are metastatic. In other embodiments, the cancer cells being treated are resistant to anticancer agents, e.g., taxanes, e.g., docetaxel.
[0069] In some embodiments, infections suitable for treatment with the methods of the present invention include, but are not limited to, infections caused by viruses, bacteria, fungi, parasites, mycoplasma, prions, and the like.
[0070] The present invention contemplates that any known therapeutic utility of gossypol may be exploited via pulsatile dose administration of gossypol. In certain embodiments, gossypol administered by pulsatile doing can be used to modulate spermicidal activity (e.g., function as a male contraceptive or antifertility agent), treat malaria, microbial or viral disease (e.g., inhibit the growth of the HIV virus as a treatment for AIDS), treat obesity, skin disorders or baldness, inhibit growth of endothelial cells, inhibit vascularization or neovascularization, treat arthritic conditions, neovascular-based dermatological conditions, diabetic retinopathy, Kaposi's sarcoma, age-related macular degeneration, restenosis, telangiectasia, glaucoma, keloids, corneal graft rejection, wound granularization, angiofibroma, Osier- Webber syndrome, myocardial angiogenesis, psoriatic arthritis or scleroderma, inhibit DNA synthesis or DNA polymerase activity and treat gynecological disorders (e.g., endometriosis) or diabetic complications.
[0071] Some embodiments of the present invention provides methods for administering an effective amount of gossypol by pulsatile dosing and at least one additional therapeutic agent (including, but not limited to, chemotherapeutic agents, antineoplastic agents, antimicrobial agents, antiviral agents, antifungal agents, and anti-inflammatory agents) and/or therapeutic technique (e.g., surgical intervention, and/or radiotherapeutic agent). The term "chemotherapeutic agent," as used herein, refers to any chemical substance known to those of skill in the art to be effective for the treatment or amelioration of cancer and/or as an inducer of apoptosis.
[0072] In some embodiments, the combination of gossypol administered by pulsatile dosing and one or more therapeutic agents will have a greater effect as compared to the administration of either gossypol or therapeutic agent alone. In other embodiments, the combination of gossypol administered by pulsatile dosing and one or more therapeutic agents is expected to result in a synergistic effect (i.e., more than additive) as compared to the administration of either one alone. In some embodiments, pulsatile dose administration of gossypol results in a reduction in number and/or decreased severity of adverse events in patients. In some embodiments, the combination of gossypol administered by pulsatile dosing and one or more anticancer agents overcomes patient chemoresistance to the anticancer agent.
[0073] In a particular embodiment, the invention pertains to a method of treating, ameliorating or preventing cancer comprising administering to a patient in need thereof (-)-gossypol in combination with an anticancer agent, wherein about 40 mg to about 60 mg of said (-)-gossypol is administered twice-a-day for three consecutive days followed by eighteen consecutive days wherein said (-)-gossypol is not administered. In one embodiment, the anticancer agent is docetaxel. In one embodiment, the patient has demonstrated chemoresi stance to docetaxel treatment. In one embodiment, the cancer is prostate cancer.
[0074] A number of suitable anticancer agents are contemplated for use in the methods of the present invention. Indeed, the present invention contemplates, but is not limited to, administration of numerous anticancer agents such as: agents that induce apoptosis; polynucleotides (e.g., anti-sense, ribozymes, siRNA); polypeptides (e.g., enzymes and antibodies); biological mimetics (e.g., BH3 mimetics); agents that bind (e.g., oligomerize or complex) with a Bcl-2 family protein such as Bax; alkaloids; alkylating agents; antitumor antibiotics; antimetabolites; hormones; platinum compounds; monoclonal or polyclonal antibodies (e.g., antibodies conjugated with anticancer drugs, toxins, defensins), toxins; radionuclides; biological response modifiers (e.g., interferons (e.g., IFN-α) and interleukins (e.g., IL-2)); adoptive immunotherapy agents; hematopoietic growth factors; agents that induce tumor cell differentiation (e.g., all-trans-retinoic acid); gene therapy reagents (e.g., antisense therapy reagents and nucleotides); tumor vaccines; angiogenesis inhibitors; proteosome inhibitors: NF-KB modulators; anti-CDK compounds; HDAC inhibitors; and the like. Numerous other examples of chemotherapeutic compounds and anticancer therapies suitable for coadministration with gossypol or compositions thereof are known to those skilled in the art.
[0075] In certain embodiments, anticancer agents comprise agents that induce or stimulate apoptosis. Agents that induce apoptosis include, but are not limited to, radiation (e.g., X-rays, gamma rays, UV); kinase inhibitors (e.g., epidermal growth factor receptor (EGFR) kinase inhibitor, vascular growth factor receptor (VGFR) kinase inhibitor, fibroblast growth factor receptor (FGFR) kinase inhibitor, platelet-derived growth factor receptor (PDGFR) kinase inhibitor, and Bcr-Abl kinase inhibitors (such as GLEEVEC)); antisense molecules; antibodies (e.g., HERCEPTIN, RITUXAN, ZEVALIN, BEXXAR, and AVASTIN); anti-estrogens (e.g., raloxifene and tamoxifen); anti-androgens (e.g., flutamide, bicalutamide, finasteride, aminoglutethamide, ketoconazole, and corticosteroids); cyclooxygenase 2 (COX-2) inhibitors (e.g., celecoxib, meloxicam, NS-398, and non-steroidal anti-inflammatory drugs); anti-inflammatory drugs (e.g., butazolidin, DECADRON, DELTASONE, dexamethasone, dexamethasone intensol, DEXONE, HEXADROL, hydroxychloroquine, METICORTEN, ORADEXON, ORASONE, oxyphenbutazone, PEDIAPRED, phenylbutazone, PLAQUENIL, prednisolone, prednisone, PRELONE, and TANDEARIL); and cancer chemotherapeutic drugs (e.g., irinotecan (CAMPTOSAR), CPT-I l, fludarabine (FLUDARA), dacarbazine, dexamethasone, mitoxantrone, MYLOTARG, VP- 16, cisplatin, carboplatin, oxaliplatin, 5-FU, doxorubicin, gemcitabine, bortezomib, gefitinib, bevacizumab, TAXOTERE or TAXOL); cellular signaling molecules; ceramides and cytokines; staurosporine; and ara- C, and the like.
[0076] In still other embodiments, the methods of the present invention provide pulsatile dose administration of gossypol and at least one anti- hyperproliferative or antineoplastic agent; e.g., selected from alkylating agents, antimetabolites, and natural products (e.g., herbs and other plant and/or animal derived compounds).
[0077] Alkylating agents suitable for use in the present compositions and methods include, but are not limited to: 1) nitrogen mustards (e.g., mechlorethamine, cyclophosphamide, ifosfamide, melphalan (L-sarcolysin); and chlorambucil); 2) ethylenimines and methylmelamines (e.g., hexamethylmelamine and thiotepa); 3) alkyl sulfonates (e.g., busulfan); 4) nitrosoureas (e.g., carmustine (BCNU); lomustine (CCNU); semustine (methyl-CCNU); and streptozocin (streptozotocin)); and 5) triazenes (e.g., dacarbazine (dimethyltriazenoimid-azolecarboxamide) .
[0078] In some embodiments, antimetabolites suitable for use in the present compositions and methods include, but are not limited to: 1) folic acid analogs (e.g., methotrexate (amethopterin)); 2) pyrimidine analogs (e.g., fluorouracil (5-fluorouracil), floxuridine (fluorode-oxyuridine), and cytarabine (cytosine arabinoside)); and 3) purine analogs (e.g., mercaptopurine (6-mercaptopurine), thioguanine (6-thioguanine), and pentostatin (2'-deoxycoformycin)).
[0079] In still further embodiments, chemotherapeutic agents suitable for use in the compositions and methods of the present invention include, but are not limited to: 1) vinca alkaloids (e.g., vinblastine, vincristine); 2) epipodophyllotoxins (e.g., etoposide and teniposide); 3) antibiotics (e.g., dactinomycin (actinomycin D), daunorubicin (daunomycin; rubidomycin), doxorubicin, bleomycin, plicamycin (mithramycin), and mitomycin (mitomycin C)); 4) enzymes (e.g., L-asparaginase); 5) biological response modifiers (e.g., interferon-alfa); 6) platinum coordinating complexes (e.g., cisplatin and carboplatin); 7) anthracenediones (e.g., mitoxantrone); 8) substituted ureas (e.g., hydroxyurea); 9) methylhydrazine derivatives (e.g., procarbazine (N-methylhydrazine)); 10) adrenocortical suppressants (e.g., mitotane (o,p'-DDD) and aminoglutethimide); 11) adrenocorticosteroids (e.g., prednisone); 12) progestins (e.g., hydroxyprogesterone caproate, medroxyprogesterone acetate, and megestrol acetate); 13) estrogens (e.g., diethylstilbestrol and ethinyl estradiol); 14) antiestrogens (e.g., tamoxifen); 15) androgens (e.g., testosterone propionate and fluoxymesterone); 16) antiandrogens (e.g., flutamide): and 17) gonadotropin-releasing hormone analogs (e.g., leuprolide).
[0080] Any oncolytic agent that is used in a cancer therapy context finds use in the methods of the present invention. For example, the U.S. Food and Drug Administration maintains a formulary of oncolytic agents approved for use in the United States. International counterpart agencies to the U.S.F.D.A. maintain similar formularies. Table 1 provides a list of exemplary antineoplastic agents approved for use in the U.S. Those skilled in the art will appreciate that the "product labels" required on all U.S. approved chemotherapeutics describe approved indications, dosing information, toxicity data, and the like, for the exemplary agents. Table 1
Figure imgf000034_0001
Figure imgf000035_0001
Figure imgf000036_0001
Figure imgf000037_0001
Figure imgf000038_0001
Figure imgf000039_0001
Figure imgf000040_0001
Figure imgf000041_0001
Figure imgf000042_0001
Figure imgf000043_0001
Anticancer agents further include compounds which have been identified to have anticancer activity but are not currently approved by the U.S. Food and Drug Administration or other counterpart agencies or are undergoing evaluation for new uses. Examples include, but are not limited to, 3-AP, 12-O-tetradecanoylphorbol-13-acetate, 17AAG, 852 A, ABI-007, ABR- 217620, ABT-751, ADI-PEG 20, AE-941, AG-013736, AGROlOO, alanosine, AMG 706, antibody G250, antineoplastons, AP23573, apaziquone, APC8015, atiprimod, ATN-161, atrasenten, azacitidine, BB-10901, BCX-1777, bevacizumab, BGOOOOl, bicalutamide, BMS 247550, bortezomib, bryostatin- 1, buserelin, calcitriol, CCI-779, CDB-2914, cefixime, cetuximab, CG0070, cilengitide, clofarabine, combretastatin A4 phosphate, CP-675,206, CP- 724,714, CpG 7909, curcumin, decitabine, DENSPM, doxercalciferol, E7070, E7389, ecteinascidin 743, efaproxiral, eflornithine, EKB-569, enzastaurin, erlotinib, exisulind, fenretinide, flavopiridol, fludarabine, flutamide, fotemustine, FR901228, Gl 7DT, galiximab, gefitinib, genistein, glufosfamide, GTI-2040, histrelin, HKI-272, homoharringtonine, HSPPC-96, hul4.18- interleukin-2 fusion protein, HuMax-CD4, iloprost, imiquimod, infliximab, interleukin-12, IPI-504, irofulven, ixabepilone, lapatinib, lestaurtinib, leuprolide, LMB-9 immunotoxin, lonafarnib, luniliximab, mafosfamide, MB07133, MDX-010, MLN2704, monoclonal antibody 3F8, monoclonal antibody J591, motexafin, MS-275, MVA-MUC1-IL2, nilutamide, nitrocamptothecin, nolatrexed dihydrochloride, nolvadex, NS-9, 06- benzylguanine, oblimersen sodium, ONYX-015, oregovomab, OSI-774, panitumumab, paraplatin, PD-0325901, pemetrexed, PHY906, pioglitazone, pirfenidone, pixantrone, PS-341, PSC 833, PXDlOl, pyrazoloacridine, Rl 15777, RADOOl, ranpirnase, rebeccamycin analogue, rhuAngiostatin protein, rhuMab 2C4, rosiglitazone, rubitecan, S-I, S-8184, satraplatin, SB-, 15992, SGN-0010, SGN-40, sorafenib, SR31747A, ST1571, SUOl 1248, suberoylanilide hydroxamic acid, suramin, talabostat, talampanel, tariquidar, temsirolimus, TGFa-PE38 immunotoxin, thalidomide, thymalfasin, tipifarnib, tirapazamine, TLK286, trabectedin, trimetrexate glucuronate, TroVax, UCN- 1, valproic acid, vinflunine, VNP40101M, volociximab, vorinostat, VX-680, ZDl 839, ZD6474, zileuton, and zosuquidar trihydrochloride.
[0082] For a more detailed description of anticancer agents and other therapeutic agents, those skilled in the art are referred to any number of instructive manuals including, but not limited to, the Physician's Desk Reference and to Goodman and Gilman's "Pharmaceutical Basis of Therapeutics" ninth edition, Eds. Hardman et al, 1996.
[0083] In one embodiment, the anticancer agent is selected from the group consisting of abraxane, actinomycin D, aldesleukin, alemtuzumab, alitretinoin, allopurinol, altretamine, amifostine, aminoglutethamide, anastrozole, arsenic trioxide, asparaginase, azacitidine, azathioprine, BCG live, bevacizumab, bexarotene, bicalutamide, bleomycin, bortezomib, busulfan, butazolidin, capecitabine, carboplatin, carmustine, celecoxib, chlorambucil, cisplatin, cladribine, clofarabine, cyclophosphamide, cytarabine, dacarbazine, dactinomycin, darbepoetin alfa, daunomycin, daunorubicin, denileukin diftitox, dexamethasone, dexrazoxane, diethylstilbestrol, docetaxel, doxorubicin, dromostanolone propionate, epirubicin, epoetin alfa, estramustine, ethinyl estradiol, etoposide, exemestane, filgrastim, finasteride, floxuridine, fludarabine, fluorouracil, fluoxymesterone, flutamide, fulvestrant, gefitinib, gemcitabine, gemtuzumab, goserelin acetate, hexamethylmelamine, hydroxychloroquine, hydroxyprogesterone caproate, hydroxyurea, ibritumomab, idarubicin, ifosfamide, imatinib, interferon alfa-2a, interferon alfa-2b, interleukin-2, irinotecan, ketoconazole, letrozole, leucovorin, leuprolide, levamisole HCl, lomustine, mechlorethamine, medroxyprogesterone acetate, megestrol acetate, meloxicam, melphalan, mercaptopurine, mesna, methotrexate, methoxsalen, methylprednisolone, metronidazole, misonidazole, mithramycin, mitomycin, mitotane, mitoxantrone, nandrolone phenpropionate, nitrogen mustard, nitroimidazole, nitrosourea, nofetumomab, oblimersen sodium, oprelvekin, oxaliplatin, oxaliplatin, oxyphenbutazone, paclitaxel, pamidronate, pegademase, pegaspargase, pegfϊlgrastim, pentostatin, phenylbutazone, picoplatin, pipobroman, plicamycin, plicamycin, porfimer sodium, prednisolone, prednisone, procarbazine, procarbazine, quinacrine, raloxifene, rasburicase, rituximab, romidepsin, sargramostim, semustine, streptozocin, talc, tamoxifen, temozolomide, teniposide, testolactone, testosterone propionate, thalidomide, thioguanine, thiotepa, tiripazamine, topotecan HCl, toremifene, tositumomab, trastuzumab, tretinoin, trimethoprim/sulfamethoxazole, uracil mustard, valrubicin, vinblastine, vincristine, vindesine, vinorelbine and zoledronic acid.
[0084] In another embodiment, the anticancer agent is a taxane. In another embodiment, the taxane is selected from the group consisting of docetaxel and paclitaxel. In another embodiment, the taxane is docetaxel.
[0085] The present invention provides methods for pulsatile dose administration of gossypol with radiation therapy. The term "radiotherapeutic agent," as used herein refers any type of radiation therapy known to those of skill in the art to be effective for the treatment or amelioration of cancer and/or as an inducer of apoptosis. The invention is not limited by the types, amounts, or delivery and administration systems used to deliver the therapeutic dose of radiation to the patient. For example, the patient may receive photon radiotherapy, particle beam radiation therapy, radioisotope therapy (e.g., radioconjugates with monoclonal antibodies), other types of radiotherapies, and combinations thereof. In some embodiments, the radiation is delivered to the patient using a linear accelerator. In still other embodiments, the radiation is delivered using a gamma knife.
[0086] The source of radiation can be external or internal to the patient.
External radiation therapy is most common and involves directing a beam of high-energy radiation to a tumor site through the skin using, for instance, a linear accelerator. While the beam of radiation is localized to the tumor site, it is nearly impossible to avoid exposure of normal, healthy tissue. However, external radiation is usually well tolerated by patients. Internal radiation therapy involves implanting a radiation-emitting source, such as beads, wires, pellets, capsules, particles, and the like, inside the body at or near the tumor site including the use of delivery systems that specifically target cancer cells (e.g., using particles attached to cancer cell binding ligands). Such implants can be removed following treatment, or left in the body inactive. Types of internal radiation therapy include, but are not limited to, brachytherapy, interstitial irradiation, intracavity irradiation, radioimmunotherapy, and the like.
[0087] The patient may optionally receive radiosensitizers (e.g., metronidazole, misonidazole, intra-arterial Budr, intravenous iododeoxyuridine (IudR), nitroimidazole, 5-substituted-4-nitroimidazoles, 2H- isoindolediones, [[(2-bromoethyl)-amino]methyl]-nitro-lH-imidazole-l- ethanol, nitroaniline derivatives, DNA-affϊnic hypoxia selective cytotoxins, halogenated DNA ligand, 1,2,4 benzotriazine oxides, 2-nitroimidazole derivatives, fluorine-containing nitroazole derivatives, benzamide, nicotinamide, acridine-intercalator, 5-thiotretrazole derivative, 3-nitro-l,2,4- triazole, 4,5-dinitroimidazole derivative, hydroxylated texaphrins, cisplatin, mitomycin, tiripazamine, nitrosourea, mercaptopurine, methotrexate, fluorouracil, bleomycin, vincristine, carboplatin, epirubicin, doxorubicin, cyclophosphamide, vindesine, etoposide, paclitaxel, heat (hyperthermia), and the like), radioprotectors (e.g., cysteamine, aminoalkyl dihydrogen phosphorothioates, amifostine (WR 2721), IL-I, IL-6, and the like). Radiosensitizers enhance the killing of tumor cells. Radioprotectors protect healthy tissue from the harmful effects of radiation.
[0088] Any type of radiation can be administered to a patient, so long as the dose of radiation is tolerated by the patient without unacceptable negative side-effects. Suitable types of radiotherapy include, for example, ionizing (electromagnetic) radiotherapy (e.g., X-rays or gamma rays) or particle beam radiation therapy (e.g., high linear energy radiation). Ionizing radiation is defined as radiation comprising particles or photons that have sufficient energy to produce ionization, i.e., gain or loss of electrons (as described in, for example, U.S. 5,770,581 incorporated herein by reference in its entirety). The effects of radiation can be at least partially controlled by the clinician. The dose of radiation is preferably fractionated for maximal target cell exposure and reduced toxicity.
[0089] The total dose of radiation administered to an animal preferably is about .01 Gray (Gy) to about 100 Gy. More preferably, about 10 Gy to about 65 Gy (e.g., about 15 Gy, 20 Gy, 25 Gy, 30 Gy, 35 Gy, 40 Gy, 45 Gy, 50 Gy, 55 Gy, or 60 Gy) are administered over the course of treatment. While in some embodiments a complete dose of radiation can be administered over the course of one day, the total dose is ideally fractionated and administered over several days. Desirably, radiotherapy is administered over the course of at least about 3 days, e.g., at least 5, 7, 10, 14, 17, 21, 25, 28, 32, 35, 38, 42, 46, 52, or 56 days (about 1-8 weeks). Accordingly, a daily dose of radiation will comprise approximately 1-5 Gy (e.g., about 1 Gy, 1.5 Gy, 1.8 Gy, 2 Gy, 2.5 Gy, 2.8 Gy, 3 Gy, 3.2 Gy, 3.5 Gy, 3.8 Gy, 4 Gy, 4.2 Gy, or 4.5 Gy), preferably 1-2 Gy (e.g., 1.5-2 Gy). The daily dose of radiation should be sufficient to induce destruction of the targeted cells. If stretched over a period, radiation preferably is not administered every day, thereby allowing the animal to rest and the effects of the therapy to be realized. For example, radiation desirably is administered on 5 consecutive days, and not administered on 2 days, for each week of treatment, thereby allowing 2 days of rest per week. However, radiation can be administered 1 day/week, 2 days/week, 3 days/week, 4 days/week, 5 days/week, 6 days/week, or all 7 days/week, depending on the animal's responsiveness and any potential side effects. Radiation therapy can be initiated at any time in the therapeutic period. Preferably, radiation is initiated in week 1 or week 2, and is administered for the remaining duration of the therapeutic period. For example, radiation is administered in weeks 1-6 or in weeks 2-6 of a therapeutic period comprising 6 weeks for treating, for instance, a solid tumor. Alternatively, radiation is administered in weeks 1-5 or weeks 2-5 of a therapeutic period comprising 5 weeks. These exemplary radiotherapy administration schedules are not intended, however, to limit the present invention.
[0090] Antimicrobial therapeutic agents may also be used as therapeutic agents in the present invention. Any agent that can kill, inhibit, or otherwise attenuate the function of microbial organisms may be used, as well as any agent contemplated to have such activities. Antimicrobial agents include, but are not limited to, natural and synthetic antibiotics, antibodies, inhibitory proteins (e.g., defensins), antisense nucleic acids, membrane disruptive agents and the like, used alone or in combination. Indeed, any type of antibiotic may be used including, but not limited to, antibacterial agents, antiviral agents, antifungal agents, and the like.
[0091] In some embodiments of the present invention, gossypol and one or more therapeutic agents are administered to an animal under one or more of the following conditions: at different periodicities, at different durations, at different concentrations, by different administration routes, etc. provided that gossypol is administered according to a discontinuous dosing regimen (i.e., via pulsatile dosing). In some embodiments, gossypol is administered by pulsatile dosing prior to the therapeutic agent, e.g., 0.5, 1, 2, 3, 4, 5, 10, 12, or 18 hours, 1, 2, 3, 4, 5, or 6 days, 1, 2, 3, or 4 weeks prior to the administration of the therapeutic agent. In some embodiments, gossypol is administered by pulsatile dosing after the therapeutic agent, e.g., 0.5, 1, 2 3, 4, 5, 10, 12, or 18 hours, 1, 2, 3, 4, 5, or 6 days, 1, 2, 3, or 4 weeks after the administration of the therapeutic agent. In some embodiments, gossypol and the therapeutic agent are administered concurrently but on different schedules, e.g., gossypol is administered on at least two consecutive days followed by at least one day wherein gossypol is not administered while the therapeutic agent is administered once a week, once every two weeks, once every three weeks, or once every four weeks. In other embodiments, gossypol is administered on one day a week while the therapeutic agent is administered daily, once a week, once every two weeks, once every three weeks, or once every four weeks. In one embodiment, (-)-gossypol is administered twice-a-day for three consecutive days every 21 days, one therapeutic agent, e.g., docetaxel, is administered every 21 days, and another therapeutic agent, e.g., prednisone, is administered daily.
[0092] The above-described dose administration schedules are provided for illustrative purposes only and should not be considered limiting.
[0093] Pharmaceutical compositions may be produced by combining gossypol in a therapeutically effective amount to induce apoptosis in cells or to sensitize cells to inducers of apoptosis with a pharmaceutically acceptable carrier. Pharmaceutical compositions may comprise, for example, (±)-gossypol, (+)- gossypol, (-)-gossypol, (±)-gossypol co-crystal, (+)-gossypol co-crystal or (-)- gossypol co-crystal.
[0094] Pharmaceutical compositions useful within the scope of this invention include all compositions wherein gossypol is contained in an amount which is effective to achieve its intended purpose, e.g., to treat, ameliorate or prevent a disease, condition or disorder responsive to the induction of apoptosis. While individual needs vary, determination of optimal ranges of effective amounts of each component in a pharmaceutical composition is within the skill of the art. In one embodiment, the pharmaceutical composition comprising gossypol may be administered to patients, e.g. humans, orally at a dose totaling about 1 mg to about 1200 mg, or an equivalent dose of the pharmaceutically acceptable salt thereof, per day. In another embodiment, a total oral dose of about 5 mg to about 500 mg, about 5 mg to about 250 mg, about 5 mg to about 100 mg, or about 5 mg to about 60 mg is administered per day. In another embodiment, a total oral dose of about 90 mg to about 240 mg is administered per day. In another embodiment, a total oral dose of about 80 mg to about 200 mg is administered per day. In another embodiment, an oral dose of about 5 mg, 10 mg, 20 mg, 30 mg, 40 mg, 50 mg, 60 mg, 70 mg, 80 mg, 90 mg, 100 mg, 110 mg, 120 mg, 130 mg, 140 mg, 150 mg, 160 mg, 170 mg, 180 mg, 190 mg or 200 mg is administered two times a day. In another embodiment, an oral dose of 30 mg, 35 mg, 40 mg, 45 mg, 50 mg, 55 mg, 60 mg, 65 mg, 70 mg, 75 mg or 80 mg is administered two times a day. In another embodiment, an oral dose of about 50 mg, 60 mg, 70 mg, 80 mg, 90 mg, 100 mg, 110 mg, 120 mg, 130 mg, 140 mg, 150 mg, 160 mg, 170 mg, 180 mg, 190 mg, 200 mg, 210 mg, 220 mg, 230 mg, 240 mg, 250 mg, 260 mg, 270 mg, 280 mg, 290 mg, 300 mg, 310 mg, 320 mg, 330 mg, 340 mg, 350 mg, 360 mg, 370 mg, 380 mg, 390 mg or 400 mg is administered once a day. In another embodiment, an oral dose of 80 mg, 90 mg, 100 mg, 110 mg, 120 mg, 130 mg, 140 mg, 150 mg, 160 mg, 170 mg, 180 mg, 190 mg or 200 mg is administered once a day. In certain embodiments, the amount (e.g., dose) of gossypol administered may increase as the period of time between dosing increases, since the potential for adverse events may decrease under such circumstances. The unit oral dose may comprise from about 1 mg to about 1000 mg of the pharmaceutical composition comprising gossypol. In one embodiment, the unit oral dose may comprise from about 5 mg to about 500 mg of the pharmaceutical composition comprising gossypol. In another embodiment, the unit oral dose may comprise from about 5 mg to about 100 mg. In another embodiment, the unit oral dose may comprise from about 5 mg to about 30 mg. In another embodiment, the unit oral dose may comprise from about 30 mg to about 80 mg. In another embodiment, the unit oral dose may comprise about 40 mg to about 60 mg. The unit dose may be administered one or more times daily as one or more tablets, capsules and the like, each containing from about 1 to about 1000 mg of the pharmaceutical composition comprising gossypol, conveniently about 5 mg to about 100 mg of the composition, e.g., about 1 mg, 5 mg, 10 mg, 15 mg, 20 mg, 25 mg, 30 mg, 35 mg, 40 mg, 45 mg, 50 mg, 55 mg, 60 mg, 65 mg, 70 mg, 75 mg, 80 mg, 85 mg, 90 mg, 95 mg or 100 mg. In one embodiment, the unit oral dose is administered two times per day. In another embodiment, the unit oral dose is administered two times per day for three consecutive days.
[0096] For intramuscular injection, the dose is generally about one-half of the oral dose. For example, a suitable intramuscular dose of gossypol would be about 0.5 mg to about 500 mg. In one embodiment, the intramuscular dose would be about 0.5 mg to about 100 mg, about 0.5 to about 50 mg, about 0.5 mg to about 25 mg, or about 0.5 mg to about 15 mg.
[0097] In a topical formulation, the composition may be present at a concentration of about 0.01 to 100 mg per gram of carrier. In one embodiment, the composition is present at a concentration of about 0.07 mg/g to about 1.0 mg/g, about 0.1 to about 0.5 mg/g, or about 0.4 mg/g.
[0098] Gossypol may be administered via pulsatile dosing as part of a pharmaceutical composition containing suitable pharmaceutically acceptable carriers comprising excipients and auxiliaries which facilitate processing of the compositions into preparations which can be used pharmaceutically. Pharmaceutical preparations which can be administered orally include tablets, dragees, slow release lozenges, capsules and the like. Pharmaceutical preparations which can be administered topically include mouth rinses and mouth washes, gels, liquid suspensions, hair rinses, hair gels, shampoos and the like. Pharmaceutical preparations which can be administered rectally include suppositories and the like. Pharmaceutical preparations for administration by injection include suitable solutions and the like. Pharmaceutical preparations administered via pulsatile dosing contain from about 0.01 to about 99 percent, or from about 0.25 to about 75 percent of gossypol, together with the excipient(s). [0099] Methods of the invention may be administered to any patient which may experience the beneficial effects of such methods. Foremost among such patients are mammals, e.g., humans, although the invention is not intended to be so limited. Other patients include veterinary animals (cows, sheep, pigs, horses, dogs, cats and the like).
[00100] Gossypol and pharmaceutical compositions thereof may be administered via pulsatile dose administration by any means that achieve their intended purpose. For example, administration may be by parenteral, subcutaneous, intravenous, intramuscular, intraperitoneal, transdermal, buccal, intrathecal, intracranial, intranasal or topical routes. Alternatively, or concurrently, administration may be by the oral route. The dosage administered will be dependent upon the age, health, and weight of the recipient, kind of concurrent treatment, if any, frequency of treatment, and the nature of the effect desired.
[00101] The pharmaceutical preparations of the present invention are manufactured in a manner which is itself known, for example, by means of conventional mixing, granulating, dragee-making, dissolving, or lyophilizing processes. Thus, pharmaceutical preparations for oral use can be obtained by combining the active compounds with solid excipients, optionally grinding the resulting mixture and processing the mixture of granules, after adding suitable auxiliaries, if desired or necessary, to obtain tablets or dragee cores.
[00102] Suitable excipients are, in particular, fillers such as saccharides, for example lactose or sucrose, mannitol or sorbitol, cellulose preparations and/or calcium phosphates, for example tricalcium phosphate or calcium hydrogen phosphate, as well as binders such as starch paste, using, for example, maize starch, wheat starch, rice starch, potato starch, gelatin, tragacanth, methyl cellulose, hydroxypropylmethylcellulose, sodium carboxymethylcellulose, and/or polyvinyl pyrrolidone. If desired, disintegrating agents may be added such as the above-mentioned starches and also carboxymethyl-starch, cross- linked polyvinyl pyrrolidone, agar, or alginic acid or a salt thereof, such as sodium alginate. Auxiliaries are, above all, flow-regulating agents and lubricants, for example, silica, talc, stearic acid or salts thereof, such as magnesium stearate or calcium stearate, and/or polyethylene glycol. Dragee cores are provided with suitable coatings which, if desired, are resistant to gastric juices. For this purpose, concentrated saccharide solutions may be used, which may optionally contain gum arabic, talc, polyvinyl pyrrolidone, polyethylene glycol and/or titanium dioxide, lacquer solutions and suitable organic solvents or solvent mixtures. In order to produce coatings resistant to gastric juices, solutions of suitable cellulose preparations such as acetylcellulose phthalate or hydroxypropylmethyl-cellulose phthalate, are used. Dye stuffs or pigments may be added to the tablets or dragee coatings, for example, for identification or in order to characterize combinations of active compound doses.
[00103] Other pharmaceutical preparations which can be used orally include push-fit capsules made of gelatin, as well as soft, sealed capsules made of gelatin and a plasticizer such as glycerol or sorbitol. The push-fit capsules can contain the active compounds in the form of granules which may be mixed with fillers such as lactose, binders such as starches, and/or lubricants such as talc or magnesium stearate and, optionally, stabilizers. In soft capsules, the active compounds are preferably dissolved or suspended in suitable liquids, such as fatty oils, or liquid paraffin. In addition, stabilizers may be added.
[00104] Possible pharmaceutical preparations which can be used rectally include, for example, suppositories, which consist of a combination of one or more of the active compounds with a suppository base. Suitable suppository bases are, for example, natural or synthetic triglycerides, or paraffin hydrocarbons. In addition, it is also possible to use gelatin rectal capsules which consist of a combination of the active compounds with a base. Possible base materials include, for example, liquid triglycerides, polyethylene glycols, or paraffin hydrocarbons.
[00105] Suitable formulations for parenteral administration include aqueous solutions of the active compounds in water-soluble form, for example, water- soluble salts and alkaline solutions. In addition, suspensions of the active compounds as appropriate oily injection suspensions may be administered. Suitable lipophilic solvents or vehicles include fatty oils, for example, sesame oil, or synthetic fatty acid esters, for example, ethyl oleate or triglycerides or polyethylene glycol-400. Aqueous injection suspensions may contain substances which increase the viscosity of the suspension include, for example, sodium carboxymethyl cellulose, sorbitol, and/or dextran. Optionally, the suspension may also contain stabilizers.
[00106] The topical compositions of this invention are formulated preferably as oils, creams, lotions, ointments and the like by choice of appropriate carriers. Suitable carriers include vegetable or mineral oils, white petrolatum (white soft paraffin), branched chain fats or oils, animal fats and high molecular weight alcohol (greater than C12). The preferred carriers are those in which the active ingredient is soluble. Emulsifiers, stabilizers, humectants and antioxidants may also be included as well as agents imparting color or fragrance, if desired. Additionally, transdermal penetration enhancers can be employed in these topical formulations. Examples of such enhancers can be found in U.S. Pat. Nos. 3,989,816 and 4,444,762.
[00107] Creams are preferably formulated from a mixture of mineral oil, self- emulsifying beeswax and water in which mixture the active ingredient, dissolved in a small amount of an oil such as almond oil, is admixed. A typical example of such a cream is one which includes about 40 parts water, about 20 parts beeswax, about 40 parts mineral oil and about 1 part almond oil.
[00108] Ointments may be formulated by mixing a suspension of the active ingredient in a vegetable oil such as almond oil with warm soft paraffin and allowing the mixture to cool. A typical example of such an ointment is one which includes about 30% almond oil and about 70% white soft paraffin by weight.
[00109] Lotions may be conveniently prepared by preparing a suspension of the active ingredient in a suitable high molecular weight alcohol such as propylene glycol or polyethylene glycol. [00110] The following examples are illustrative, but not limiting, of the methods of the present invention. Suitable modifications and adaptations of the variety of conditions and parameters normally encountered in clinical therapy and which are obvious to those skilled in the art are within the spirit and scope of the invention.
EXAMPLE 1
Pulsatile Dosing of Gossypol
[00111] A phase I clinical trial was carried out to compare the maximum tolerated dose and safety of daily (i.e., continuous) versus pulsatile (i.e., intermittent) dosing of (-)-gossypol in patients with advanced cancer. A secondary objective of this study was to identify any anti-tumor activity of (-)-gossypol. Patients were treated with increasing doses of (-)-gossypol according to the following dosing schedules: "Daily" dosing: 5 to 60 mg/day of (-)-gossypol on 21 days per 28 day cycle; "BID X 3d" dosing: 30 to 80 mg BID of (-)-gossypol on 3 consecutive days (e.g., Monday-Tuesday- Wednesday) repeated every other week per 28 day cycle; and "Weekly" dosing: 80 to 200 mg of (-)-gossypol once weekly per 28 day cycle. Adverse events (AEs) were graded by NCI-CTCAE v3. Overall, pulsatile dosing (BID X 3d and Weekly) resulted in a reduced percentage of AEs, particularly Grade 3/4 AEs, as compared to continuous daily dosing (see Table 2, Any AE).
Table 2
Figure imgf000055_0001
Figure imgf000056_0001
Figure imgf000057_0001
Figure imgf000058_0001
EXAMPLE 2
Clinical Efficacy of Gossypol ] Following (-)-gossypol administration to patients with advanced cancer, clinical efficacy (e.g., patients having stable disease for 60 days or more) was monitored according to the following dosing schedules: "Daily" dosing: 5 to 60 mg/day of (-)-gossypol on 21 days per 28 day cycle; "BID X 3d" dosing: 30 to 80 mg BID of (-)-gossypol on 3 consecutive days (e.g., Monday-Tuesday- Wednesday) repeated every other week per 28 day cycle; and "Weekly" dosing: 80 to 200 mg of (-)-gossypol once weekly per 28 day cycle. Pulsatile dosing (BID X 3d) resulted in a longer median duration of days of stable disease as compared to continuous daily dosing (Table 3).
Table 3
Figure imgf000058_0002
Figure imgf000059_0001
EXAMPLE 3
In Vivo Efficacy of (-)-Gossypol Acetic Acid Co-Crystals in the A549 Non- Small Cell Cancer (NSCLC) Xenograft Model
[00113] The in vivo efficacy of (-)-gossypol acetic acid co-crystals alone or in combination with taxotere (TXT) in the A549 NSCLC xenograph model is shown in Figs. 2 and 3. About 5 million cells of A549 were inoculated into nude mice, 8 mice per dosing group. In one experiment, (-)-gossypol acetic acid co-crystals were administered at 15 mg/kg, oral dosing (po), daily for 21 days, either alone or in combination with taxotere at 8 mg/kg, iv, once a week for three weeks (Fig. 2). In another experiment, (-)-gossypol acetic acid co- crystals were administered at 60 mg/kg, po, daily for three days per week (day 1-3/week) every two weeks (days 1-3, and then days 15-17), either alone or in combination with taxotere at 30 mg/kg, iv, single dose only, once every three weeks (Fig. 3). The results of these studies show inter alia that an intermittent dosing of (-)-gossypol acetic acid co-crystals in combination with taxotere effectively reduces tumor volume.
EXAMPLE 4 Clinical Study of Gossypol in Combination with Docetaxel
[00114] An open-label non-randomized Phase I-II clinical trial (labeled CS-
202) is being carried out on patients with histologically confirmed metastatic prostate cancer. Briefly, patients are divided in into two cohorts: Cohort A- Patients with rising PSA level on hormonal therapy, i.e., hormone-refractory patients, and Cohort B-Patients with PSA or disease progression on docetaxel (taxotere), i.e., docetaxel-refractory patients. All patients were treated with 40-60 mg (-)-gossypol acetic acid co-crystal 40-60 mg BID days 1-3, every 21 days, 75 mg/m2 docetaxel q 21 days and prednisone, daily per label.
[00115] Phase I data from 9 patients is summarized as follows: no dose- limiting toxicities, no severe gastrointestinal toxicity, no increase in docetaxel hematological toxicity.
[00116] Phase I-II data is available on first 20 patients of Cohort A. The patient demographics are provided in Table 4. See N. Engl. J. Med. 357:1502- 1512 (2004) for further details of TAX 327 clinical study.
Table 4
Figure imgf000060_0001
[00117] Figure 4 is a waterfall plot illustrating the therapeutic response of the first 20 patients of Cohort A. These data indicate a high rate of PSA response with (-)-gossypol acetic acid co-crystal treatment. In addition, only one patient out of 20 was refractory at the onset of treatment Data from this study also indicates steep declines in the slope of PSA response versus time and PSA reductions occur early in the treatment and are long lasting (Figure 5). Finally, CT scanning shows tumor size reduction following treatment with (-)- gossypol, docetaxel, and prednisone (Figure 6). Table 5 shows the preliminary efficacy and safety comparisons between CS-202 and TAX 327. Patients receiving (-)-gossypol acetic acid co- crystal (labeled "(-)-gossy") show a marked improvement in 50% PSA reduction and a significantly lower refractory rate. ("D" = docetaxel; "P" = prednisone).
Table 5
Figure imgf000061_0001
Table 6 compares the PSA responses of (-)-gossypol acetic acid co- crystal Cohort A combination therapy with approved docetaxel monotherapy regimens and other investigational combination therapies for the treatment of hormone refractory prostate cancer. (-)-Gossypol acetic acid co-crystal (labeled "(-)-gossypol") responses compare favorably to those reported in these other studies.
Table 6
Figure imgf000063_0001
1-N Engl J Med 351:1502 -1512, 2004; 2 - J Clin Oncol 25:669 -674, 2007; 3 - N Engl J Med 351:1513-1520, 2004; 4 - J Natl Cancer Inst 98:516 -521, 2006; 5 - J Clin Oncol 25:3965-3970, 2007
[00120] Figure 7 illustrates the therapeutic response of the first 11 patients of
Cohort B. These data indicate the desired PSA response with (-)-gossypol acetic acid co-crystal treatment in 6 out to the 11 patients, with 4 patients having a 30+% decline and 2 patients having a 50+% decline in PSA response.
[00121] Figures 8-10 illustrates the rapid therapeutic response when
(-)-gossypol acetic acid co-crystal is added to a taxotere regimen of various patients of Cohort B. The abrupt reversal of the PSA response (from rising to falling) suggests pulsatile dosing of (-)-gossypol acetic acid co-crystal may be especially effective in treating chemoresistant patients.
[00122] Having now fully described the invention, it will be understood by those of skill in the art that the same can be performed within a wide and equivalent range of conditions, formulations, and other parameters without affecting the scope of the invention or any embodiment thereof. All patents, patent applications and publications cited herein are fully incorporated by reference herein in their entirety.

Claims

WHAT IS CLAIMED IS:
1. A method of treating, ameliorating or preventing a disease, condition or disorder responsive to treatment with gossypol comprising administering to a patient in need thereof a therapeutically effective amount of said gossypol by pulsatile dose administration.
2. The method of claim 1, wherein said disease, condition or disorder is responsive to the induction of apoptosis.
3. The method of claim 2, wherein said disease, condition or disorder responsive to the induction of apoptosis is selected from the group consisting of a hyperproliferative disease, autoimmune disorder, chronic inflammatory condition, viral infection, microbial infection, parasitic infection, osteoarthritis, and atherosclerosis.
4. The method of claim 3, wherein said chronic inflammatory condition is psoriasis.
5. The method of claim 3, wherein said disease, condition or disorder responsive to the induction of apoptosis is selected from the group consisting of viral infection, microbial infection, and parasitic infection.
6. The method of claim 3, wherein said disease, condition or disorder responsive to the induction of apoptosis is a hyperproliferative disease.
7. The method of claim 6, wherein said hyperproliferative disease is cancer.
8. The method of claim 7, wherein said cancer is selected from the group consisting of breast cancer, prostate cancer, lymphoma, skin cancer, pancreatic cancer, colon cancer, malignant melanoma, ovarian cancer, brain cancer, primary brain carcinoma, head-neck cancer, glioma, glioblastoma, liver cancer, bladder cancer, non-small cell lung cancer, head carcinoma, neck carcinoma, breast carcinoma, ovarian carcinoma, lung carcinoma, small-cell lung carcinoma, Wilms1 tumor, cervical carcinoma, testicular carcinoma, bladder carcinoma, pancreatic carcinoma, stomach carcinoma, colon carcinoma, prostatic carcinoma, genitourinary carcinoma, thyroid carcinoma, esophageal carcinoma, myeloma, multiple myeloma, adrenal carcinoma, renal cell carcinoma, endometrial carcinoma, adrenal cortex carcinoma, malignant pancreatic insulinoma, malignant carcinoid carcinoma, choriocarcinoma, mycosis fungoides, malignant hypercalcemia, cervical hyperplasia, leukemia, acute lymphocytic leukemia, chronic lymphocytic leukemia, acute myelogenous leukemia, chronic myelogenous leukemia, chronic granulocytic leukemia, acute granulocytic leukemia, hairy cell leukemia, neuroblastoma, rhabdomyosarcoma, Kaposi's sarcoma, polycythemia vera, essential thrombocytosis, Hodgkin's disease, non-Hodgkin's lymphoma, soft-tissue sarcoma, osteogenic sarcoma, primary macroglobulinemia and retinoblastoma.
9. The method of claim 1, wherein said patient suffers fewer and/or less severe adverse events compared to when said gossypol is administered daily.
10. The method of claim 1 further comprising administering one or more additional therapeutic agents.
11. The method of claim 10, wherein said disease, disorder or condition is cancer and said one or more additional therapeutic agents comprises one or more anticancer agents.
12. The method of claim 11, wherein said one or more anticancer agents is selected from the group consisting of abraxane, actinomycin D, aldesleukin, alemtuzumab, alitretinoin, allopurinol, altretamine, amifostine, aminoglutethamide, anastrozole, arsenic trioxide, asparaginase, azacitidine, azathioprine, BCG live, bevacizumab, bexarotene, bicalutamide, bleomycin, bortezomib, busulfan, butazolidin, capecitabine, carboplatin, carmustine, celecoxib, chlorambucil, cisplatin, cladribine, clofarabine, cyclophosphamide, cytarabine, dacarbazine, dactinomycin, darbepoetin alfa, daunomycin, daunorubicin, denileukin diftitox, dexamethasone, dexrazoxane, diethylstilbestrol, docetaxel, doxorubicin, dromostanolone propionate, epirubicin, epoetin alfa, estramustine, ethinyl estradiol, etoposide, exemestane, filgrastim, finasteride, fioxuridine, fludarabine, fluorouracil, fiuoxymesterone, flutamide, fulvestrant, gefitinib, gemcitabine, gemtuzumab, goserelin acetate, hexamethylmelamine, hydroxychloroquine, hydroxyprogesterone caproate, hydroxyurea, ibritumomab, idarubicin, ifosfamide, imatinib, interferon alfa-2a, interferon alfa-2b, interleukin-2, irinotecan, ketoconazole, letrozole, leucovorin, leuprolide, levamisole HCl, lomustine, mechlorethamine, medroxyprogesterone acetate, megestrol acetate, meloxicam, melphalan, mercaptopurine, mesna, methotrexate, methoxsalen, methylprednisolone, metronidazole, misonidazole, mithramycin, mitomycin, mitotane, mitoxantrone, nandrolone phenpropionate, nitrogen mustard, nitroimidazole, nitrosourea, nofetumomab, oblimersen sodium, oprelvekin, oxaliplatin, oxaliplatin, oxyphenbutazone, paclitaxel, pamidronate, pegademase, pegaspargase, pegfilgrastim, pentostatin, phenylbutazone, picoplatin, pipobroman, plicamycin, plicamycin, porfimer sodium, prednisolone, prednisone, procarbazine, procarbazine, quinacrine, raloxifene, rasburicase, rituximab, romidepsin, sargramostim, semustine, streptozocin, talc, tamoxifen, temozolomide, teniposide, testolactone, testosterone propionate, thalidomide, thioguanine, thiotepa, tiripazamine, topotecan HCl, toremifene, tositumomab, trastuzumab, tretinoin, trimethoprim/sulfamethoxazole, uracil mustard, valrubicin, vinblastine, vincristine, vindesine, vinorelbine and zoledronic acid.
13. The method of claim 12, wherein said anticancer agent is a taxane.
14. The method of claim 13, wherein said taxane is selected from the group consisting of docetaxel and paclitaxel.
15. The method of claim 10, wherein said one or more additional therapeutic agents comprises an inducer of apoptosis.
16. The method of claim 15, wherein said inducer of apoptosis is one or more chemotherapeutic agents.
17. The method of claim 15, wherein said inducer of apoptosis is one or more radiotherapeutic agents.
18. The method of claim 15, wherein said inducer of apoptosis is a combination of one or more chemotherapeutic agents and one or more radiotherapeutic agents.
19. The method of claim 11, wherein said patient is chemoresistant to said one or more anticancer agents.
20. The method of claim 19, wherein said one or more anticancer agents is a taxane.
21. The method of claim 20, wherein said taxane is docetaxel.
22. The method of claim 19, wherein said cancer is selected from the group consisting of breast cancer, prostate cancer, lymphoma, skin cancer, pancreatic cancer, colon cancer, malignant melanoma, ovarian cancer, brain cancer, primary brain carcinoma, head-neck cancer, glioma, glioblastoma, liver cancer, bladder cancer, non-small cell lung cancer, head carcinoma, neck carcinoma, breast carcinoma, ovarian carcinoma, lung carcinoma, small-cell lung carcinoma, Wilms' tumor, cervical carcinoma, testicular carcinoma, bladder carcinoma, pancreatic carcinoma, stomach carcinoma, colon carcinoma, prostatic carcinoma, genitourinary carcinoma, thyroid carcinoma, esophageal carcinoma, myeloma, multiple myeloma, adrenal carcinoma, renal cell carcinoma, endometrial carcinoma, adrenal cortex carcinoma, malignant pancreatic insulinoma, malignant carcinoid carcinoma, choriocarcinoma, mycosis fungoides, malignant hypercalcemia, cervical hyperplasia, leukemia, acute lymphocytic leukemia, chronic lymphocytic leukemia, acute myelogenous leukemia, chronic myelogenous leukemia, chronic granulocytic leukemia, acute granulocytic leukemia, hairy cell leukemia, neuroblastoma, rhabdomyosarcoma, Kaposi's sarcoma, polycythemia vera, essential thrombocytosis, Hodgkin's disease, non-Hodgkin's lymphoma, soft-tissue sarcoma, osteogenic sarcoma, primary macroglobulinemia and retinoblastoma.
23. The method of claim 22, wherein said cancer is prostate cancer.
24. The method of claim 10, wherein said gossypol is administered prior to said one or more additional therapeutic agents.
25. The method of claim 10, wherein said gossypol is administered concurrently with said one or more additional therapeutic agents.
26. The method of claim 10, wherein said gossypol is administered after said one or more additional therapeutic agents.
27. The method of claim 10, wherein said gossypol and said one or more additional therapeutic agents are administered with different periodicities, different durations, different concentrations, and different administration routes.
28. The method of claims 1 or 10, wherein said gossypol is administered no more frequently than one day out of every two days.
29. The method of claims 1 or 10, wherein said gossypol is administered on at least two consecutive days followed by at least one day wherein said gossypol is not administered.
30. The method of claim 29, wherein said gossypol is administered on at least three consecutive days.
31. The method of claim 30, wherein said gossypol is administered on three consecutive days followed by at least seven consecutive days wherein said gossypol is not administered.
32. The method of claim 31, wherein said gossypol is administered on three consecutive days followed by at least eleven consecutive days wherein said gossypol is not administered.
33. The method of claim 32, wherein said gossypol is administered on three consecutive days followed by at least seventeen consecutive days wherein said gossypol is not administered.
34. The method of claims 1 or 10, wherein said gossypol is administered on one day followed by six consecutive days wherein said gossypol is not administered.
35. The method of claim 34, wherein said administration is continued for at least three weeks.
36. The method of claims 1 or 10, wherein said method is repeated at least once.
37. The method of claims 1 or 10, wherein said method is repeated at least once after a waiting period of at least one day wherein said gossypol is not administered.
38. The method of claim 37, wherein said waiting period is seven days.
39. The method of claim 37, wherein said waiting period is fourteen days.
40. The method of claim 37, wherein said waiting period is thirty days.
41. The method of claim 1 or 10, wherein said pulsatile dose administration comprises the sequential use of a combination of two or more different pulsatile dose administration schedules.
42. The method of claim 41, wherein said schedules are separated by a waiting period of at least one day between each schedule wherein said gossypol is not administered.
43. The method of claim 42, wherein said waiting period is seven days.
44. The method of claim 42, wherein said waiting period is fourteen days.
45. The method of claim 42, wherein said waiting period is thirty days.
46. The method of claim 10, wherein a pharmaceutical composition comprising said gossypol is orally administered.
47. The method of claim 29, wherein about 5 mg to about 200 mg BID of said gossypol is orally administered.
48. The method of claim 47, wherein about 10 mg to about 150 mg of said gossypol is administered.
49. The method of claim 48, wherein about 20 mg to about 100 mg of said gossypol is administered.
50. The method of claim 30, wherein about 5 mg to about 200 mg BID of said gossypol is orally administered.
51. The method of claim 50, wherein about 10 mg to about 150 mg of said gossypol is administered.
52. The method of claim 51 , wherein about 20 mg to about 100 mg of said gossypol is administered.
53. The method of claim 52, wherein about 30 mg to about 90 mg of said gossypol is administered.
54. The method of claim 53, wherein about 40 mg to about 60 mg of gossypol is administered.
55. The method of claim 54, wherein about 40 mg of gossypol is administered.
56. The method of claim 33, wherein about 10 mg to about 1000 mg of said gossypol is orally administered.
57. The method of claim 56, wherein about 25 mg to about 500 mg of said gossypol is administered.
58. The method of claim 57, wherein about 50 mg to about 400 mg of said gossypol is administered.
59. The method of claim 58, wherein about 80 to about 300 mg of said gossypol is administered.
60. The method of claims 1 or 10, wherein said gossypol is (-)-gossypol.
61. A method of treating, ameliorating or preventing cancer comprising administering to a patient in need thereof (-)-gossypol in combination with an anticancer agent, wherein about 40 mg to about 60 mg of said (-)-gossypol is orally administered twice-a-day for three consecutive days followed by eighteen consecutive days wherein said (-)-gossypol is not administered.
62. The method of claim 61, wherein said anticancer agent is docetaxel.
63. The method of claim 61, wherein said patient has demonstrated chemoresistance to said anticancer agent.
64. The method of claim 63, wherein said anticancer agent is docetaxel.
65. The method of claims 62 or 64, wherein said cancer is prostate cancer.
66. The method of claim 61, wherein about 40 mg of said (-)-gossypol is administered and said anticancer agent is docetaxel, wherein about 75 mg/m2 of said docetaxel is intravenously administered every 21 days.
67. The method of claim 66 further comprising daily administration of prednisone.
PCT/US2008/006936 2007-05-31 2008-06-02 Pulsatile dosing of gossypol for treatment of disease WO2008150506A1 (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
AU2008260510A AU2008260510A1 (en) 2007-05-31 2008-06-02 Pulsatile dosing of gossypol for treatment of disease
EP08768032A EP2152076A4 (en) 2007-05-31 2008-06-02 Pulsatile dosing of gossypol for treatment of disease
CA002689033A CA2689033A1 (en) 2007-05-31 2008-06-02 Pulsatile dosing of gossypol for treatment of disease
JP2010510372A JP2010529023A (en) 2007-05-31 2008-06-02 Gossypol's rhythmic dosing for the treatment of disease

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US94121707P 2007-05-31 2007-05-31
US60/941,217 2007-05-31

Publications (1)

Publication Number Publication Date
WO2008150506A1 true WO2008150506A1 (en) 2008-12-11

Family

ID=40094025

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2008/006936 WO2008150506A1 (en) 2007-05-31 2008-06-02 Pulsatile dosing of gossypol for treatment of disease

Country Status (6)

Country Link
US (1) US20090010878A1 (en)
EP (1) EP2152076A4 (en)
JP (1) JP2010529023A (en)
AU (1) AU2008260510A1 (en)
CA (1) CA2689033A1 (en)
WO (1) WO2008150506A1 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2016135732A1 (en) * 2015-02-26 2016-09-01 Yeda Research And Development Co. Ltd. Method of promoting hair growth
CN109942455A (en) * 2019-03-10 2019-06-28 陕西科技大学 Gossypol with anti-tumor activity-Eflornithine schiff base compounds and its synthetic method

Families Citing this family (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB0011903D0 (en) * 2000-05-18 2000-07-05 Astrazeneca Ab Combination chemotherapy
US8173686B2 (en) 2006-11-06 2012-05-08 Poniard Pharmaceuticals, Inc. Use of picoplatin to treat colorectal cancer
US8178564B2 (en) * 2006-11-06 2012-05-15 Poniard Pharmaceuticals, Inc. Use of picoplatin to treat colorectal cancer
US8168661B2 (en) * 2006-11-06 2012-05-01 Poniard Pharmaceuticals, Inc. Use of picoplatin to treat colorectal cancer
US8168662B1 (en) 2006-11-06 2012-05-01 Poniard Pharmaceuticals, Inc. Use of picoplatin to treat colorectal cancer
US20110033528A1 (en) * 2009-08-05 2011-02-10 Poniard Pharmaceuticals, Inc. Stabilized picoplatin oral dosage form
US20100260832A1 (en) * 2007-06-27 2010-10-14 Poniard Pharmaceuticals, Inc. Combination therapy for ovarian cancer
TW200916094A (en) * 2007-06-27 2009-04-16 Poniard Pharmaceuticals Inc Stabilized picoplatin dosage form
CN101809024A (en) * 2007-07-16 2010-08-18 铂雅制药公司 The oral preparations of pyrrole platinum
US7696372B2 (en) * 2007-10-01 2010-04-13 Ascenta Therapeutics, Inc. Process for preparing R-gossypol L-phenylalaninol dienamine
US20110052581A1 (en) * 2008-02-08 2011-03-03 Poniard Pharmaceuticals Inc. Use of picoplatin and cetuximab to treat colorectal cancer
EP2449894B1 (en) 2009-07-02 2018-08-15 Japan Tobacco, Inc. Tobacco product to be used in oral cavity
WO2013153821A1 (en) * 2012-04-12 2013-10-17 Omura Satoshi Pdk4 inhibitor and use thereof
KR101579371B1 (en) * 2014-02-27 2015-12-22 국립암센터 Anti-cancer composition comprising gossypol and phenformin
KR101765575B1 (en) 2015-02-02 2017-08-07 연세대학교 산학협력단 Pharmaceutical composition for inhibiting a growth of cancer stem cells comprising aldehyde inhibitor and biguanide compounds
EP3417853B1 (en) 2016-02-18 2024-06-05 Industry-Academic Cooperation Foundation Yonsei University Pharmaceutical composition for treatment of stomach cancer containing gossypol, phenformin and irinotecan
IL268224B2 (en) * 2017-02-22 2024-04-01 Nat Cancer Ct Pharmaceutical composition for preventing and treating pancreatic cancer, containing gossypol and phenformin as active ingredients
KR102428313B1 (en) * 2018-07-09 2022-08-03 연세대학교 산학협력단 A pharmaceutical composition for the treatment of brain cancer comprising aldehyde inhibitor, and an anticancer agent
CN109438323B (en) * 2018-11-13 2022-03-11 陕西盘龙药业集团股份有限公司 Gossypol-7-N heteroisatin Schiff base compounds with anti-tumor activity and synthesis method thereof
CN109575050B (en) * 2018-11-13 2022-03-11 陕西盘龙药业集团股份有限公司 Gossypol-7-N heteroisatin Schiff base compounds with anti-tumor activity and synthesis method thereof

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030105030A1 (en) * 1997-10-31 2003-06-05 Shutsung Liao Method and compositions for regulation of 5-alpha reductase activity
US20040214902A1 (en) * 2001-05-30 2004-10-28 The Regents Of The University Of Michigan Small molecule antagonists of BCL-2 family proteins

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5385936A (en) * 1990-07-12 1995-01-31 The United States Of America As Represented By The Secretary Of The Department Of The Health And Human Services Gossypol acetic acid for the treatment of cancer
EA011218B1 (en) * 2004-03-25 2009-02-27 Дзе Риджентс Оф Дзе Юниверсити Оф Мичиган Gossypol co-crystals and use thereof

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030105030A1 (en) * 1997-10-31 2003-06-05 Shutsung Liao Method and compositions for regulation of 5-alpha reductase activity
US20040214902A1 (en) * 2001-05-30 2004-10-28 The Regents Of The University Of Michigan Small molecule antagonists of BCL-2 family proteins

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See also references of EP2152076A4 *

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2016135732A1 (en) * 2015-02-26 2016-09-01 Yeda Research And Development Co. Ltd. Method of promoting hair growth
CN109942455A (en) * 2019-03-10 2019-06-28 陕西科技大学 Gossypol with anti-tumor activity-Eflornithine schiff base compounds and its synthetic method

Also Published As

Publication number Publication date
EP2152076A1 (en) 2010-02-17
EP2152076A4 (en) 2011-02-16
AU2008260510A1 (en) 2008-12-11
US20090010878A1 (en) 2009-01-08
JP2010529023A (en) 2010-08-26
CA2689033A1 (en) 2008-12-11

Similar Documents

Publication Publication Date Title
EP2152076A1 (en) Pulsatile dosing of gossypol for treatment of disease
US9745314B2 (en) MDM2 inhibitors and therapeutic methods using the same
US20100189683A1 (en) Pulsatile Dosing of Gossypol for Treatment of Disease
EP1732529A1 (en) Gossypol co-crystals and the use thereof
EP2707372B1 (en) Spiro-oxindole mdm2 antagonists
WO2006050447A2 (en) Apogossypolone and the uses thereof
US20060247305A1 (en) Chromen-4-one inhibitors of anti-apoptotic Bcl-2 family members and the uses thereof
EP1853242A2 (en) Small molecule inhibitors of stat3 and the uses thereof
WO2010056932A1 (en) Pulsatile dosing of gossypol for treatment of disease
EP2007706A2 (en) Production of gossypol co-crystals
WO2016085990A1 (en) Compositions and methods relating to inhibiting serine hyrdoxymethyltransferase 2 activity
WO2017031116A1 (en) Small molecule inhibitors of ku70/80 and uses thereof
US10781183B2 (en) Small molecule inducers of reactive oxygen species and inhibitors of mitochondrial activity
WO2021011778A1 (en) Small molecule inhibitors of ku70/80 and uses thereof
EP3630083A1 (en) Dimethyl-nonatetraenyl-trimethyl-cyclohexyl compounds and uses thereof

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: 08768032

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 581252

Country of ref document: NZ

WWE Wipo information: entry into national phase

Ref document number: 2008260510

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 2010510372

Country of ref document: JP

Ref document number: 2008768032

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2689033

Country of ref document: CA

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2008260510

Country of ref document: AU

Date of ref document: 20080602

Kind code of ref document: A