WO2009108857A2 - Combination therapy for prostate cancer - Google Patents

Combination therapy for prostate cancer Download PDF

Info

Publication number
WO2009108857A2
WO2009108857A2 PCT/US2009/035464 US2009035464W WO2009108857A2 WO 2009108857 A2 WO2009108857 A2 WO 2009108857A2 US 2009035464 W US2009035464 W US 2009035464W WO 2009108857 A2 WO2009108857 A2 WO 2009108857A2
Authority
WO
WIPO (PCT)
Prior art keywords
inhibitor
prostate cancer
combination
igf
egcg
Prior art date
Application number
PCT/US2009/035464
Other languages
French (fr)
Other versions
WO2009108857A3 (en
Inventor
Peter Kushner
Cyrus Harmon
Leslie Hodges-Gallagher
William Stafford
Original Assignee
Combithera, 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 Combithera, Inc. filed Critical Combithera, Inc.
Publication of WO2009108857A2 publication Critical patent/WO2009108857A2/en
Publication of WO2009108857A3 publication Critical patent/WO2009108857A3/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/4738Quinolines; Isoquinolines ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/4745Quinolines; Isoquinolines ortho- or peri-condensed with heterocyclic ring systems condensed with ring systems having nitrogen as a ring hetero atom, e.g. phenantrolines

Definitions

  • Prostate cancer is frequently detected at an early stage using the prostate specific antigen (PSA) test and subsequent biopsy. It is treated most often either with surgical removal of the prostate or by destruction of the prostate with radiation or other means. Men with early stage prostate cancer also elect “watchful waiting” in which surgery is delayed or suspended until the cancer worsens. In addition there are substantial numbers of men with pre-cancerous conditions of the prostate such as prostatic intraepithelial neoplasia (PIN) on watchful waiting because these conditions frequently progress to cancer.
  • PIN prostatic intraepithelial neoplasia
  • prostate cancer can recur, and is often detected by an increase in prostate specific antigen (PSA) after the initial post-surgical decline.
  • PSA prostate specific antigen
  • Recurrent prostate cancer is often treated with androgen ablation therapy, an effective treatment for this stage of the disease where growth of tumor cells is dependent on androgen stimulation.
  • the response to androgen ablation is limited as the tumor progresses to androgen independence via mutations that make proliferation of the tumor cells independent of the action of the androgen receptor.
  • Histone deacetylase (HDAC) inhibitors are a structurally diverse group of pharmacological agents that inhibit proliferation, induce differentiation and/or apoptosis in a wide range of cancer cells and hold much promise as anti-neoplastic agents (Villar- Garea, A. and M. Esteller, Int J Cancer, 2004. 112(2): p. 171-8; Marks, P., et al, Nat Rev Cancer, 2001. 1(3): p. 194-202).
  • Hyperacetylation is associated with a transcriptionally permissive environment and HDAC inhibitors, although they affect only a small number of target genes, activate genes involved in cell cycle arrest, apoptosis and differentiation (Glaser, K.B., et al, MoI Cancer Ther, 2003. 2(2): p. 151-63; Richon, V.M., et al, Proc Natl Acad Sci USA, 2000. 97(18): p. 10014-9; Munster, P.N., et al., Cancer Res., 2001. 61(23): p. 8492-7).
  • HDAC inhibitors increase the efficiency of several anticancer drugs that target the DNA (Kim, M.S., et al., Cancer Res, 2003. 63(21): p. 7291-300; Castro-Galache, M.D., et al., Int J Cancer, 2003. 104(5): p. 579-86).
  • a variety of small molecule HDAC inhibitors are currently in preclinical development.
  • the present inventors have investigated the ability of cocktails of targeted anticancer agents, including HDAC inhibitors, to inhibit the proliferation of cell lines derived from various stages of prostate cancer; androgen dependent, androgen independent (albeit AR positive), and completely androgen independent (AR negative).
  • HDAC inhibitors include HDAC inhibitors, androgen dependent, androgen independent (albeit AR positive), and completely androgen independent (AR negative).
  • the inventors have discovered that, surprisingly, certain HDAC inhibitors combine effectively with other therapies for prostate cancer cells at particular stages, and that the combinations do not reduce the activity of any single component, and that these combinations are more effective than either of the components alone.
  • HDAC inhibitors including VPA, sulforaphane (SFN), and trichostatin (TSA) in combination with the mTOR inhibitor rapamycin, the IGF-IR inhibitor picropodophyllin (PPP), and the green tea polyphenol, epigallocatechin gallate (EGCG), also an IGF-IR inhibitor, as well as androgen ablation therapy, such as bicalutamide.
  • VPA sulforaphane
  • TSA trichostatin
  • the present inventors have found that for certain stages of prostate cancer, certain combinations of HDAC inhibitors and one or more other compounds, including, but not limited to IGF-IR inhibitors, mTOR inhibitors and androgen ablation therapy combine effectively with each other and are superior to other combinations of compounds for the treatment and prevention of prostate cancer, and for preventing the progression of prostate cancer.
  • the data provided herein show that certain agents that inhibit prostate cancer cell proliferation on their own, combine effectively for prostate cancer cells from certain stages of cancer but, surprisingly, not for others (for example, in androgen-dependent prostate cancer cells, such as LNCaP cells versus androgen independent prostate cancer cells, such as DU145 cells).
  • the present inventors have found that the combination of VPA and EGCG (both of which inhibit prostate cancer cell proliferation as single agents) was effective on earlier stage (androgen- dependent) prostate cancer cells, but not on later stage (androgen-independent) prostate cancer cells.
  • the combination of the HDAC inhibitor, SFN and the mTOR inhibitor, rapamycin is effective against later stage (androgen- independent) prostate cancer cells.
  • the present invention provides methods of treating and preventing prostate cancer at various stages, including androgen dependent, androgen independent but androgen receptor (AR) positive, and completely androgen independent (AR negative) prostate cancer using the combinations that are effective for the particular stage of cancer, as well as pharmaceutical compositions comprising the compounds used in the combination therapies.
  • AR androgen dependent, androgen independent but androgen receptor
  • AR negative completely androgen independent
  • the present invention encompasses methods of treatment for (including management of, amelioration of symptoms of, and preventing the progression of) prostate cancer, using certain combination therapies, as well as the pharmaceutical compositions comprising these combination therapies.
  • the invention is based, in part, on the recognition that certain combinations of compounds combine effectively with each other, and are superior to other combinations of compounds, as well as improving the tolerance of, and/or reducing the side effects caused by at least one of the compounds in the combination.
  • Subjects are mammalian, and preferably are human, and more preferably are human males.
  • the present invention encompasses methods of treating and methods of preventing prostate cancer comprising administering to a subject suffering therefrom a therapeutically effective amount of an HDAC inhibitor in combination with a therapeutically effective amount of one or more of an IGF-I receptor inhibitor, and/or an mTOR inhibitor, and optionally, androgen ablation therapy.
  • various combinations of these compounds, or a single one of the compounds is administered in combination with androgen ablation therapy.
  • one or more of the compounds are administered in combination with an anti-androgen, such as bicalutamide.
  • the present invention also encompasses methods and compositions for preventing prostate cancer, particularly in subjects who are at risk for prostate cancer that is greater than the average risk for prostate cancer.
  • Risk factors considered in preventing prostate cancer in subjects include, but are not limited to those diagnosed with prostatic intraepithelial neoplasia (PIN), prostate-specific antigen (PSA) levels, family history of prostate cancer (relatives with prostate cancer), age, age at which relatives were diagnosed, circulating hormone levels, genetic markers for prostate cancer such as PCA3 and 8q24, diet, obesity, race, ethnicity, nationality, whether the individual has had a vasectomy, and presence of prostatitis.
  • the present invention also provides methods and compositions for preventing the progression of prostate cancer to a later stage for those who already have prostate cancer or precancerous indicators, as well as preventing the recurrence of prostate cancer for those in remission from prostate cancer.
  • the invention contemplates methods of preventing the progression of prostatic intraepithelial neoplasia (PIN) and methods of preventing the progression of PIN to prostate cancer.
  • the invention encompasses treating or preventing androgen (AR)-dependent prostate cancer, in some preferred embodiments, the invention encompasses treating or preventing androgen independent (albeit AR positive) prostate cancer, and in yet other preferred embodiments, the invention encompasses treating or preventing completely androgen independent (AR negative) prostate cancer.
  • the present invention encompasses treating PIN, including low grade (PIN I) and high grade (PIN II and PIN III) lesions.
  • the present invention encompasses methods of treating and methods of preventing prostate cancer comprising administering to a subject suffering therefrom a therapeutically effective amount of an HDAC inhibitor in combination with a therapeutically effective amount of one or more of an IGF-I receptor inhibitor, or an mTOR inhibitor, in combination with androgen ablation therapy, in individuals with androgen-dependent prostate cancer.
  • one or more of the compounds are administered in combination with bicalutamide.
  • the present invention also provides pharmaceutical compositions comprising or consisting essentially of the combinations of compounds described herein, as well as kits comprising the combinations.
  • the HDAC inhibitors encompassed by the present invention can be any known to those of skill in the art, including those described in Minucci et al, Nature 6:38- 51 (2006). According to the invention, in some embodiments, the HDAC inhibitor is VPA. In other embodiments, the HDAC inhibitor may be SFN or TSA. In yet other embodiments, the HDAC inhibitor may be SAHA or any other HDAC inhibitor known by the skilled practitioner to be effective.
  • the patient is treated with a combination of an HDAC inhibitor, and one or more of an IGF-IR inhibitor, and/or one or more of an mTOR inhibitor.
  • the combination also comprises androgen ablation therapy.
  • the patient is treated with a combination of an HDAC inhibitor and an IGF-IR inhibitor.
  • the patient is treated with a combination of an HDAC inhibitor and an mTOR inhibitor.
  • the combination also comprises androgen ablation therapy.
  • a patient with AR-responsive, early-stage prostate cancer is treated with a combination of VPA and EGCG.
  • a patient with AR-responsive, early-stage prostate cancer is treated with VPA, EGCG, and rapamycin.
  • one or more of the compounds are administered in combination with the bicalutamide. In other embodiments, these combinations are used to prevent the progression from pre-cancerous stages to prostate cancer in patients.
  • a patient with AR-independent prostate cancer, and particularly AR-independent prostate cancer which is still AR-positive is treated with a combination of SFN and EGCG.
  • a patient with AR-independent prostate cancer, and in particular, AR-independent prostate cancer which is still AR- positive is treated with a combination of SFN and PPP.
  • a patient with AR-independent prostate cancer is treated with a combination of SFN and rapamycin.
  • a patient is treated with a combination of an HDAC inhibitor, an IGF-IR inhibitor, and an mTOR inhibitor.
  • the patient is treated with rapamycin, PPP, and SFN.
  • a patient with AR-responsive prostate cancer is treated with bicalutamide and VPA.
  • a patient with AR-responsive prostate cancer is treated with bicalutamide, VPA, and EGCG.
  • methods of preventing progression from PIN to prostate cancer comprise administering a combination of an HDAC inhibitor, and one or more of an IGF-IR inhibitor, and/or one or more of an mTOR inhibitor.
  • the combination also comprises androgen ablation therapy.
  • methods of slowing progression of na ⁇ ve (not yet treated with androgen ablation therapy) recurrent prostate cancer comprise administering a combination of an HDAC inhibitor, and one or more of an IGF-IR inhibitor, and/or one or more of an mTOR inhibitor.
  • the combination also comprises androgen ablation therapy.
  • methods of preventing resistance to androgen ablation therapy comprise administering a combination of an HDAC inhibitor, and one or more of an IGF-IR inhibitor, and/or one or more of an mTOR inhibitor, in addition to androgen ablation therapy.
  • the dose of valproic acid is between about 2g/day and about 3g/day.
  • the daily dose of valproic acid is from about 15/mg/kg to about 60 mg/kg.
  • the daily dose of valproic acid is sufficient to achieve about 300 to about 867 micromolar in patient serum. In another embodiment, the daily dose of valproic acid is sufficient to achieve about 300 to about 1000 micromolar in patient serum, and in another embodiment, the daily dose of valproic acid is sufficient to achieve about 500 to about 1000 micromolar in patient serum. [0030] In some embodiments, the dose of SAHA is from about 200 mg/day to about 600 mg/day. In another embodiment, the dose of SAHA is about 400 mg/day.
  • compounds are administered in combination, with ratios of those compounds which preserve the recommended daily doses of the compounds. In some embodiments, compounds are administered in combination, with ratios of those compounds which preserve the ranges of doses as described herein.
  • the present invention further contemplates that the combination therapies as described herein can also reduce or eliminate other side effects of treatment, at least in part because lower doses of compounds can be used in treatment or prevention protocols.
  • the IGF-IR inhibitor can be picropodophyllin (see, e.g., Girnita, A. et al, Cancer Res., 2004. 64(1): 236-242) or the green tea polyphenol, EGCG (see, e.g., Shimizu, M. et al, Biochem. Biophys. Res. Commun., 2005. 334(3): 947-953; Li, M. et al., Cancer Epidemiol. Biomarkers Prev., 2007. 16(3): 598- 605.
  • the mTOR inhibitor may be rapamycin or rapamycin derivatives (see, e.g., Johnston, S.R., Clin. Cancer Res., 2006. 12(3 Pt. 2): 1061-1069s).
  • the skilled practitioner will be able to use a variety of IGF-IR and mTOR inhibitors in the invention, to provide therapeutically effective combinations with various HDAC inhibitors.
  • the dose of EGCG is from about 800 mg/day to about 1200 mg/day.
  • the dose of EGCG is from about 300 mg/day to about 800 mg/day.
  • the dose of rapamycin is from about 0.125 mg/day to about 1 mg/day.
  • the combination comprises a ratio of VPA: EGCG from about 2 to about 3 parts VPA to about 1 part EGCG.
  • an additional "active ingredient" that can be used in combination with combination therapies described above.
  • the additional active ingredient a other chemotherapeutic agent, biologic, radiation therapy, or other agents and procedures useful in the treatment of cancer, including androgen ablation therapy.
  • the HDAC inhibitor is administered on a daily basis, while the IGF- IR inhibitor and/or mTOR inhibitor is administered every other day.
  • the IGF-IR inhibitor and/or mTOR inhibitor is administered on a daily basis and the HDAC inhibitor is administered every other day.
  • the invention also contemplates administering the HDAC inhibitor and the -IR inhibitor and/or mTOR inhibitor concurrently.
  • prostate cancer that is androgen- dependent, androgen-independent but AR positive, and androgen-independent.
  • the invention also contemplates treating subjects with prostate cancer for whom previous therapy has failed, or for whom the cancer is recurring.
  • the invention contemplates treating subjects who are genetically predisposed to prostate cancer or otherwise at increased risk.
  • the invention also encompasses methods of treating subjects to prevent progression of prostate cancer, and in some embodiments, the invention encompasses treating or preventing prostate cancer in patients with precancerous growths or benign tumors. It is within the scope of the invention to treat subjects that are in remission from prostate cancer, and to treat subjects with prostate cancer that have previously undergone treatment.
  • FIGS. 1A-1C The effect of the HDAC inhibitors, SFN, TSA, and VPA alone or in combination with rapamycin, further in combination with either the IGF-IR inhibitor, EGCG or PPP on cell proliferation of the human prostate cancer cell line, LNCaP after being in culture for 6 days.
  • Cells were treated as indicated with the following doses of inhibitors: 500 ⁇ M VPA, 2nM TSA, 1 ⁇ M SFN, 7 ⁇ M EGCG, 3OnM PPP, 2nM rapamycin.
  • Asterisks denote that a combination of drugs produces a more efficacious response than any of the drugs used alone.
  • FIGS. 2A-2C The effect of the HDAC inhibitors, SFN, TSA, and VPA alone or in combination with rapamycin, further in combination with either the IGF-IR inhibitor, EGCG or PPP on cell proliferation of the human prostate cancer cell line, DU145 after being in culture for 6 days.
  • Cells were treated as indicated with the following doses of inhibitors: 500 ⁇ M VPA, 2nM TSA, 1 ⁇ M SFN, 7 ⁇ M EGCG, 3OnM PPP, 2nM rapamycin.
  • Asterisks denote that a combination of drugs produces a more efficacious response than any of the drugs used alone.
  • FIGS. 3A-3C The effect of the HDAC inhibitors, SFN, TSA, and VPA alone or in combination with rapamycin, further in combination with either the IGF-IR inhibitor, EGCG or PPP on cell proliferation of the human prostate cancer cell line, PC3 after being in culture for 6 days.
  • Cells were treated as indicated with the following doses of inhibitors: 500 ⁇ M VPA, 2nM TSA, 1 ⁇ M SFN, 7 ⁇ M EGCG, 3OnM PPP, 2nM rapamycin.
  • FIGS. 4A-4B A) Dose response of the anti-androgen, bicalutamide in androgen-sensitive prostate cells. LNCaP cells were treated with the indicated doses of bicalutamide in the presence of 5OnM androstenediol. B) The effects of bicalutamide, the HDAC inhibitor, VPA, and the IGF-IR inhibitor EGCG, alone and in double or triple combination on cell proliferation of the human prostate cancer cell line, LNCaP after being in culture for 6 days.
  • HDAC inhibitors and other compounds including, but not limited to IGF-IR inhibitors, and mTOR inhibitors combine effectively with each other for the treatment and prevention of prostate cancer, and for preventing the progression of prostate cancer, as well as for preventing the progression of prostatic intraepithelial neoplasia (PIN).
  • PIN prostatic intraepithelial neoplasia
  • the present invention provides methods of treating and preventing prostate cancer, as well as pharmaceutical compositions comprising the compounds used in the combination therapies disclosed herein.
  • the invention also provides combinations of HDAC inhibitors, including but not limited to SFN, TSA, SAHA, and VPA, as well as inhibitors of the mammalian target of rapamycin (mTOR) protein such as rapamycin or derivatives thereof, and inhibitors of the insulin-like growth factor receptor (IGF-IR) signaling pathway such as picropodophyllin and EGCG.
  • HDAC inhibitors including but not limited to SFN, TSA, SAHA, and VPA
  • mTOR mammalian target of rapamycin
  • IGF-IR insulin-like growth factor receptor
  • the combination of HDAC inhibitor, IGF-IR inhibitor, and mTOR inhibitor can also be used, as well as other combinations as described herein, including combinations with androgen ablation therapy, such as bicalutamide.
  • Each of these combinations can further be used in combination with other therapies to treat prostate cancer, as described herein.
  • the individual compounds in the combination therapies combine effectively with each other, and in other embodiments, the individual compounds in the combination therapies synergize with each other.
  • cancer refers to a disease involving cells that have the potential to metastasize to distal sites and exhibit phenotypic traits that differ from those of non-cancer cells. Cancer cells acquire a characteristic set of functional capabilities during their development, albeit through various mechanisms. Such capabilities include evading apoptosis, self-sufficiency in growth signals, insensitivity to anti-growth signals, tissue invasion/metastasis, limitless replicative potential, and sustained angiogenesis.
  • cancer cell is meant to encompass both pre -malignant and malignant cancer cells.
  • histone deacetylase inhibitor and “inhibitor of histone deacetylase” mean a compound which is capable of interacting with a histone deacetylase and inhibiting its enzymatic activity.
  • “Inhibiting histone deacetylase enzymatic activity” means reducing the ability of a histone deacetylase to remove an acetyl group from a histone. (see, e.g., FIG. 8 and Minucci et al, Nature 6:38-51 (2006). In some preferred embodiments, such reduction of histone deacetylase activity is at least about 50%, more preferably at least about 75%, and still more preferably at least about 90%.
  • histone deacetylase activity is reduced by at least 95% and more preferably by at least 99%.
  • Assays for determining inhibition are described in Phiel, C. J., et al., J Biol Chem., 2001. 276(39): p. 36734-41 and Gottlich, M., et al., Embo J., 2001. 20(24): p. 6969-78.
  • the histone deacetylase inhibitor reduces the ability of a histone deacetylase to remove an acetyl group from a histone at a concentration that is lower than the concentration of the inhibitor that is required to produce another, unrelated biological effect.
  • the concentration of the inhibitor required for histone deacetylase inhibitory activity is at least 2-fold lower, more preferably at least 5 -fold lower, even more preferably at least 10-fold lower, and most preferably at least 20-fold lower than the concentration required to produce an unrelated biological effect.
  • active ingredient includes having a therapeutic or prophylactic effect on prostate cancer in the combinations. This does not include inactive ingredients such as pharmaceutical carriers, excipients, and the like.
  • rapamycin protein inhibitor or "mTOR inhibitor” includes drugs such as rapamycin, temsirolimus, and everolimus that selectively inhibit the mammalian target of rapamycin (mTOR).
  • IGF-I receptor inhibitor refers to drugs such as picrophodophyllin and podophyllotoxin that selectively inhibit the IGF-I receptor.
  • “Androgen ablation therapy” refers to any type of therapy, including drug treatment, procedures, or surgery, which reduces the amount of androgen in an individual.
  • the terms "prevent,” “preventing” and “prevention” refer to the prevention of the recurrence, worsening, or spread of a disease in a subject resulting from the administration of a prophylactic or therapeutic agent.
  • overexpress refers to a protein or nucleic acid (RNA) that is translated or transcribed at a detectably greater level, usually in a cancer cell, in comparison to a normal cell.
  • the term includes overexpression due to transcription, post transcriptional processing, translation, post-translational processing, cellular localization (e.g., organelle, cytoplasm, nucleus, cell surface), and RNA and protein stability, as compared to a normal cell.
  • Overexpression can be detected using conventional techniques for detecting mRNA (i.e., RT-PCR, PCR, hybridization, microarray) or proteins (i.e., ELISA, immunohistochemical techniques).
  • Overexpression can be 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or more in comparison to a normal cell. In certain instances, overexpression is 1-fold, 2-fold, 3 -fold, 4-fold or more higher levels of transcription or translation in comparison to a normal cell.
  • the term “in combination” refers to the use of more than one prophylactic and/or therapeutic agents.
  • the use of the term “in combination” does not restrict the order in which prophylactic and/or therapeutic agents are administered to a subject with cancer, especially prostate cancer.
  • a first prophylactic or therapeutic agent can be administered prior to (e.g., 1 minute, 5 minutes, 15 minutes, 30 minutes, 45 minutes, 1 hour, 2 hours, 4 hours, 6 hours, 12 hours, 24 hours, 48 hours, 72 hours, 96 hours, 1 week, 2 weeks, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 8 weeks, or 12 weeks before), concomitantly with, or subsequent to (e.g., 1 minute, 5 minutes, 15 minutes, 30 minutes, 45 minutes, 1 hour, 2 hours, 4 hours, 6 hours, 12 hours, 24 hours, 48 hours, hours, 96 hours, 1 week, 2 weeks, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 8 weeks, or 12 weeks after) the administration of a second prophylactic or therapeutic agent to a subject which had, has, or is susceptible to cancer, especially prostate cancer.
  • the prophylactic or therapeutic agents are administered to a subject in a sequence and within a time interval such that the agent of the invention can act together with the other agent to provide an increased benefit than if they were administered otherwise. Any additional prophylactic or therapeutic agent can be administered in any order with the other additional prophylactic or therapeutic agents.
  • the term “combine effectively” refers to a combination of therapies (e.g., a combination of prophylactic or therapeutic agents) which is more effective than any single agent administered alone. Combining effectively may also refer to combinations of therapies that are not less effective than any single agent or even less effective than any single agent, but which also eliminate or reduce the adverse effects of one or more of the agents.
  • the term "synergistic” refers to a combination of therapies (e.g., a combination of prophylactic or therapeutic agents) which is more effective than the additive effects of any two or more single agents.
  • a synergistic effect of a combination of therapies permits the use of lower dosages of one or more of the therapies and/or less frequent administration of said therapies (e.g., agents) to a subject with a disease or disorder, in particular, cancer, or a condition or symptom associated therewith.
  • a synergistic effect can result in improved efficacy of therapies in the prevention, management, or treatment of a disease or disorder, in particular, cancer or a condition or symptom associated therewith.
  • the synergistic effect of a combination of therapies may avoid or reduce adverse or unwanted side effects associated with the use of any single therapy. [0060]
  • side effects encompasses unwanted and adverse effects of a prophylactic or therapeutic agent.
  • Adverse effects are always unwanted, but unwanted effects are not necessarily adverse.
  • An adverse effect from a prophylactic or therapeutic agent might be harmful or uncomfortable or risky.
  • Side effects from chemotherapy include, but are not limited to, gastrointestinal toxicity such as, but not limited to, early and late-forming diarrhea and flatulence, nausea, vomiting, anorexia, leukopenia, anemia, neutropenia, asthenia, abdominal cramping, fever, pain, loss of body weight, dehydration, alopecia, dyspnea, insomnia, dizziness, mucositis, xerostomia, and kidney failure, as well as constipation, nerve and muscle effects, temporary or permanent damage to kidneys and bladder, flu-like symptoms, fluid retention, and temporary or permanent infertility.
  • Side effects from radiation therapy include but are not limited to fatigue, dry mouth, and loss of appetite.
  • Side effects from biological therapies/immunotherapies include but are not limited to rashes or swellings at the site of administration, flu-like symptoms such as fever, chills and fatigue, digestive tract problems and allergic reactions.
  • Side effects from hormonal therapies include but are not limited to nausea, fertility problems, depression, loss of appetite, eye problems, headache, and weight fluctuation. Additional undesired effects typically experienced by patients are numerous and known in the art. Many are described in the Physicians' Desk Reference (56 th ed., 2002).
  • therapeutically effective amount or dose or “therapeutically sufficient amount or dose” or “effective or sufficient amount or dose” herein is meant a dose that produces therapeutic effects for which it is administered, in the context of the combination therapy in which it is administered.
  • therapeutically effective or sufficient amount or dose of the compounds comprising the pharmaceutical compositions of the invention will be lower when administered in the specific combinations, than the doses that would be therapeutically effective or sufficient when the compounds are administered separately.
  • the exact dose will depend on the purpose of the treatment, and will be ascertainable by one skilled in the art using known techniques (see, e.g., Lieberman, Pharmaceutical Dosage Forms (vols.
  • a therapeutically effective amount refers to that amount of the therapeutic agent sufficient to destroy, modify, control or remove primary, regional or metastatic cancer tissue.
  • a therapeutically effective amount may refer to the amount of therapeutic agent sufficient to delay or minimize the spread of cancer.
  • a therapeutically effective amount may also refer to the amount of the therapeutic agent that provides a therapeutic benefit in the treatment or management of cancer.
  • a therapeutically effective amount with respect to a therapeutic agent of the invention means that amount of therapeutic agent alone, or in combination with other therapies, that provides a therapeutic benefit in the treatment or management of cancer.
  • the therapeutically effective dose can often be lower than the conventional therapeutically effective dose for non-sensitized cells.
  • a therapeutically effective amount refers to the amount of a therapeutic agent that, e.g., reduces the proliferation of cancer cells, increases the death of cancer cells or, reduces the size of a tumor or spread of a tumor in a subject.
  • a therapeutically effective amount of a therapeutic agent reduces the size of a tumor or the spread of a tumor in a subject 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%, ate 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 99% relative to a control such as PBS.
  • a therapeutically effective amount refers to the amount of a therapeutic agent that increases survival by 1 month, 2 months, 6 months, 1 year, 2 years, 3 years, 4 years, 5 years, 6 years, 7 years, 8 years, 9 years, 10 years, or more. In some embodiments, a therapeutically effective amount refers to the amount of a therapeutic agent that prevents the progression from PIN to prostate cancer.
  • the HDAC inhibitors encompassed by the methods and compositions of the present invention can be any known to those of skill in the art, including those described in Minucci et al, N ' ature 6:38-51 (2006).
  • the HDAC inhibitor is SFN, and in other embodiments the HDAC inhibitor is valproic acid.
  • the HDAC inhibitor may be TSA, SAHA, VPA derivatives, MS-275, clyclic hydroxamic acid-containing peptide, Apicidin, Trapoxin, or other HDAC inhibitors known by the skilled practitioner to be effective.
  • Other HDAC inhibitors encompassed by the methods and compositions of the invention include the VPA derivatives as described in U.S.
  • HDAC inhibitors encompassed by the methods and compositions of the invention include the sulfonyl derivatives as described in U.S. Patent No. 7,205,304 to Van Emelen, the alpha-ketoepoxide compounds of U.S.
  • Patent No. 7,057,057 to Lan- Hargest the HDAC inhibitors based on trihalomethylcarbonyl compounds as described in U.S. Patent No. 7,193,105 to Lan-Hargest, and the HDAC inhibitors based on alpha- chalcogenmethylcarbonyl compounds of U.S. Patent No. 7,214,831 to Lan-Hargest.
  • Each of the above patents and patent application publications is hereby incorporated by reference in its entirety.
  • the present invention provides a method of treating and methods of preventing prostate cancer, comprising administering to a subject a therapeutically effective amount of an HDAC inhibitor in combination with a therapeutically effective amount of an IGF-IR inhibitor, and/or an mTOR innhibitor.
  • Any of the therapies may be further combined with androgen ablation therapy, such as bicalutamide.
  • androgen ablation therapy such as bicalutamide.
  • HDAC inhibitors and IGF-IR inhibitors, and/or mTOR inhibitors, as well as types of androgen ablation therapy are contemplated as useful in treating prostate cancer.
  • the daily dose of valproic acid is from about 15/mg/kg to about 60 mg/kg.
  • the dose of valproic acid is sufficient to achieve from about 300 to about 1000 micromolar in patient serum. In another embodiment, the dose of valproic acid is sufficient to achieve from about 300 to about 867 micromolar in patient serum. In another embodiment, the dose of valproic acid is sufficient to achieve from about 500 to about 1000 micromolar in patient serum.
  • the dose of SAHA is from about 200 mg/day to about 600 mg/day. In another embodiment, the dose of SAHA is about 400 mg/day.
  • compounds are administered in combination, with ratios of those compounds which preserve the recommended daily doses of the compounds. In some embodiments, compounds are administered in combination, with ratios of those compounds which preserve the ranges of doses as described herein.
  • the present invention also encompasses methods of treating and methods of preventing prostate cancer or PIN, methods of preventing the progression from PIN to prostate cancer, methods of preventing progression from low to higher grade prostate cancer, and methods of preventing recurrence of prostate cancer, comprising administering to a subject suffering therefrom a subject a therapeutically effective amount of an HDAC inhibitor in combination with a therapeutically effective amount of one or more of an IGF-IR inhibitor, and/or an mTOR inhibitor and optionally further in combination with androgen ablation therapy.
  • a preferred combination is VPA and EGCG for the treatment of pre-cancerous or early stage, androgen dependent prostate cancer.
  • the invention further contemplates methods of slowing the progression of na ⁇ ve (not yet treated with androgen ablation therapy) recurrent prostate cancer, comprising administering to a subject suffering therefrom a subject a therapeutically effective amount of an HDAC inhibitor in combination with a therapeutically effective amount of one or more of an IGF-IR inhibitor, and/or an mTOR inhibitor.
  • the invention further contemplates methods of preventing resistance to androgen ablation therapy, comprising administering to a subject suffering therefrom a subject a therapeutically effective amount of an HDAC inhibitor in combination with a therapeutically effective amount of one or more of an IGF-IR inhibitor, and/or an mTOR inhibitor, in addition to androgen ablation therapy.
  • the combination comprises SFN, PPP, and rapamycin.
  • the combination for treating a patient with AR-responsive prostate cancer comprises VPA and EGCG.
  • the combination for treating a patient with AR independent (either AR positive or AR negative) prostate cancer comprises SFN and EGCG.
  • the combination for treating a patient with AR independent (either AR positive or AR negative) prostate cancer comprises SFN and PPP.
  • the combination for treating a patient with AR independent (either AR positive or AR negative) prostate cancer comprises SFN and rapamycin.
  • the combination for treating a patient with AR independent (either AR positive or AR negative) prostate cancer comprises EGCG, PPP and mTOR.
  • the IGF-IR inhibitors contemplated in the methods and compositions of the present invention can be any known to one of skill in the art.
  • the IGF-IR inhibitor may be for example, the green tea polyphenol, epigallocatechin gallate (EGCG), picropodophyllin (PPP), podophyllotoxin, podophyllotoxin derivatives, including those disclosed in U.S. Patent Application No. 20070123491 to Axelson, cyclolignans such as those disclosed in U.S. Patent Application No. 2004/0186169 to Larsson, and IGF-IR inhibitors such as those disclosed in U.S. Patent Application No. 20060193772 to Ochiai.
  • EGCG epigallocatechin gallate
  • PPP picropodophyllin
  • podophyllotoxin podophyllotoxin derivatives
  • cyclolignans such as those disclosed in U.S. Patent Application No. 2004/0186169 to Larsson
  • IGF-IR inhibitors such as those disclosed in U.S. Patent Application No. 20060193772 to Ochiai.
  • the mTOR inhibitors contemplated in the methods and compositions of the present invention can be any known to one of skill in the art.
  • the mTOR inhibitor may be for example, temsirolimus, everolimus, rapamycin and rapamycin derivatives, including those rapamycin derivatives disclosed in U.S. Patent Application No. 20040147541 to Lane, which is hereby incorporated by reference in its entirety.
  • the combination comprises SFN and rapamycin.
  • the combination comprises TSA and rapamycin.
  • the combination comprises VPA and rapamycin.
  • the combination comprises SFN and EGCG. In another embodiment, the combination comprises TSA and EGCG. In yet another embodiment, the combination comprises VPA and EGCG.
  • the combination comprises rapamycin and EGCG.
  • the combination comprises SFN, rapamycin, and EGCG.
  • the combination comprises TSA, rapamycin, and EGCG.
  • the combination comprises VPA, rapamycin, and EGCG.
  • the combination comprises SFN and PPP. In another embodiment, the combination comprises TSA and PPP. In another embodiment, the combination comprises VPA and PPP. In another embodiment, the combination comprises rapamycin and PPP. In another embodiment, the combination comprises SFN, rapamycin, and PPP. In yet another embodiment, the combination comprises TSA, rapamycin, and PPP. In another embodiment, the combination comprises VPA, rapamycin, and PPP. [0080] In yet another embodiment, the combination comprises bicalutamide and VPA.
  • the dose of rapamycin is from about 0.125 mg/day to about 1 mg/day.
  • the dose of valproic acid is between about 2g/day and about 3g/day.
  • the daily dose of valproic acid is from about 15/mg/kg to about 60 mg/kg.
  • the daily dose of valproic acid is sufficient to achieve about 300 to about 867 micromolar in patient serum. In another embodiment, the daily dose of valproic acid is sufficient to achieve about 300 to about 1000 micromolar in patient serum, and in another embodiment, the daily dose of valproic acid is sufficient to achieve about 500 to about 1000 micromolar in patient serum.
  • the dose of SAHA is from about 200 mg/day to about 600 mg/day. In another embodiment, the dose of SAHA is about 400 mg/day.
  • the dose of EGCG is from about 800 mg/day to about 1200 mg/day.
  • the dose of EGCG is from about 300 mg/day to about 800 mg/day.
  • the dose of rapamycin is from about 0.125 mg/day to about 1 mg/day.
  • the combination comprises a ratio of VPA: EGCG from about 2 to about 3 parts VPA to about 1 part EGCG.
  • inhibitors described herein many different procedures can be used to specifically inactivate or silence a target gene or inhibit the activity of its gene product, as encompassed by the present invention. Inhibition of protein activity can be brought about at the level of gene transcription, protein translation or post-translational modifications. For instance, the activity of a protein can be inhibited by directly inhibiting the activity of the protein such as altering a catalytic domain or alternatively by reducing the amount of the protein in the cell by reducing the amount of mRNA encoding the protein. In each case, the level of protein activity in the cell is reduced.
  • Various techniques can be used to knock down the activity of a protein and these include knockout technologies (antibodies, antisense RNA, and RNA interference) and compounds that specifically inhibit the protein activity.
  • RNA interference (RNAi) molecule is used to decrease expression of a gene.
  • RNA interference (RNAi) is defined as the ability of double-stranded RNA (dsRNA) to suppress the expression of a gene corresponding to its own sequence.
  • dsRNA double-stranded RNA
  • PTGS post-transcriptional gene silencing
  • the antisense strand of the fragment separates enough from the sense strand so that it hybridizes with the complementary sense sequence on a molecule of endogenous cellular mRNA. This hybridization triggers cutting of the mRNA in the double-stranded region, thus destroying its ability to be translated into a polypeptide. Introducing dsRNA corresponding to a particular gene thus knocks out the cell's own expression of that gene in particular tissues and/or at a chosen time.
  • Double-stranded (ds) RNA can be used to interfere with gene expression in mammals (Wianny & Zemicka-Goetz, 2000, Nature Cell Biology 2: 70-75; incorporated herein by reference in its entirety).
  • dsRNA is used as inhibitory RNA or RNAi of the function of the gene of interest to produce a phenotype that is the same as that of a null mutant of the gene of interest (Wianny & Zernicka-Goetz, 2000, Nature Cell Biology 2: 70-75).
  • Any therapy e.g., chemotherapies, radiation therapies, hormonal therapies, and/or biological therapies/immunotherapies
  • chemotherapies e.g., radiation therapies, hormonal therapies, and/or biological therapies/immunotherapies
  • Any therapy e.g., radiation therapies, hormonal therapies, and/or biological therapies/immunotherapies
  • the anti-cancer agents contemplated in the methods and compositions of the present invention which can be administered in combination with the compositions of the present invention include, but are not limited to suramin, estramustine, vinblastine, estramustine, etoposide, estramustine, Taxol, interferon, liarozole, prednisone, mitoxantrone, Paclitaxel, Docetaxel, Adriamycin, Provenge, and hormone-deprivation therapy.
  • the subject is a mammal.
  • Preferred mammals include primates such as humans and chimpanzees, domestic animals such, as horses, cows, pigs, etc. and pets such as dogs and cats.
  • the invention encompasses treating humans, and in particular, human males.
  • the pharmaceutical compositions described herein may be used for the treatment of cancer, particularly for prostate cancer.
  • the pharmaceutical compositions and methods of the present invention can be used to treat an individual with any type and/or stage of prostate cancer. There are several types of prostate cancer and there are several stages of prostate cancer, all of which are contemplated as treated by the methods and compositions of the present invention.
  • stage I the tumor is so small that it cannot be felt by digital rectal examination (DRE).
  • stage II the tumor is larger, and can be felt by DRE, but has not spread beyond the prostate.
  • stage III the cancer has spread to the local tissue near the prostate.
  • stage IV the cancer has spread to the lymph nodes or has metastasized to other body areas.
  • Clinical staging estimates how much cancer there is based on the results of the physical exam, imaging tests (x-rays, CT scans, etc.) and sometimes biopsies of affected areas. For certain cancers the results of other tests, such as blood tests, are also used in staging.
  • Pathologic staging can only be done on patients who have had surgery to remove or explore the extent of the cancer. It combines the results of clinical staging (physical exam, imaging tests, etc.) with the results from the surgery. In some cases, the pathologic stage may be different from the clinical stage (for example, if the surgery shows the cancer is more extensive than it was previously thought to be). Restaging is sometimes used to determine the extent of the disease if a cancer recurs (comes back) after treatment.
  • the methods and compositions of the present invention are used to treat patients with stage I prostate cancer. [0099] In one embodiment, the methods and compositions of the present invention are used to treat patients with stage II prostate cancer.
  • the methods and compositions of the present invention are used to treat patients with stage III prostate cancer.
  • the methods and compositions of the present invention are used to treat patients with stage IV prostate cancer, i.e. patients with metastatic cancer.
  • the patient having prostate cancer has already failed other treatment regimens such as chemotherapy.
  • the methods and pharmaceutical compositions of the present invention may be used to prevent the development of a cancer, particularly in an individual at higher risk than average to develop such cancer than other individuals, or to treat a patient afflicted with prostate cancer.
  • the invention provides methods treating asymptomatic patients who have a likelihood of benefiting from therapeutic treatment of prostate cancer.
  • the asymptomatic patients can comprise patients in any of the many high risk groups for prostate cancer.
  • Risk factors considered in preventing prostate cancer in subjects include, but are not limited to those diagnosed with prostatic intraepithelial neoplasia (PIN), prostate-specific antigen (PSA) levels, family history of prostate cancer (relatives with prostate cancer), age, age at which relatives were diagnosed, circulating hormone levels, genetic markers for prostate cancer such as PCA3 and 8q24, prostate stem cell antigen (PCSA), polymorphisms within the androgen receptor gene, levels of plasma hormones and sex hormone-binding globulin (SHBG), diet, obesity, race, ethnicity, nationality, whether the individual has had a vasectomy, and presence of prostatitis.
  • PIN prostatic intraepithelial neoplasia
  • PSA prostate-specific antigen
  • family history of prostate cancer restores with prostate
  • the present invention also provides methods and compositions for preventing the progression of prostate cancer to a later stage for those who already have prostate cancer or precancerous indicators, as well as preventing the recurrence of prostate cancer for those in remission from prostate cancer.
  • the combinations of the invention may be administered either prior to or following surgical removal of primary tumors and/or treatment such as administration of radiotherapy, surgery, or conventional chemotherapeutic drugs.
  • the methods and compositions of the present invention may be used advantageously in combination with any other treatment regimen for prostate cancer.
  • Treatments for prostate cancer are well known in the art and continue to be developed. Treatments include but are not limited to surgery, including cryosurgery and radical surgery (prostatectomy), radiation therapy, transperineal brachytherapy, and chemotherapy. Any combination of therapies may be used in conjunction with the present invention.
  • the methods and compositions comprising the combination therapies described herein may also be used to reduce the proliferation of cancer cells, increase the death of cancer cells or, reduces the size of a tumor or spread of a tumor in a subject. It is contemplated by the present invention that the combination therapies described herein may reduce the size of a tumor or the spread of a tumor in a subject 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 99% relative to a control such as PBS.
  • the combination therapies described herein may increase survival by 1 month, 2 months, 6 months, 1 year, 2 years, 3 years, 4 years, 5 years, 6 years, 7 years, 8 years, 9 years, 10 years, or more, it may render the subject disease-free, or it may prevent the progression from PIN pr other precancerous stages to prostate cancer.
  • compositions of the present invention are determined in part by the particular composition being administered, as well as by the particular method used to administer the composition. Accordingly, there are a wide variety of suitable formulations of pharmaceutical compositions of the present invention (see, e.g., Remington 's Pharmaceutical Sciences, 20 th ed., 2003).
  • the compounds of the invention may be formulated into pharmaceutical compositions as natural or salt forms.
  • Pharmaceutically acceptable non-toxic salts include the base addition salts (formed with free carboxyl or other anionic groups) which may be derived from inorganic bases such as, for example, sodium, potassium, ammonium, calcium, or ferric hydroxides, and such organic bases as isopropylamine, trimethylamine, 2-ethylamino-ethanol, histidine, procaine, and the like.
  • Such salts may also be formed as acid addition salts with any free cationic groups and will generally be formed with inorganic acids such as, for example, hydrochloric, sulfuric, or phosphoric acids, or organic acids such as acetic, p-toluenesulfonic, methanesulfonic acid, oxalic, tartaric, mandelic, and the like.
  • Salts of the invention include amine salts formed by the protonation of an amino group with inorganic acids such as hydrochloric acid, hydrobromic acid, hydroiodic acid, sulfuric acid, phosphoric acid, and the like.
  • Salts of the invention also include amine salts formed by the protonation of an amino group with suitable organic acids, such as p-toluenesulfonic acid, acetic acid, and the like.
  • suitable organic acids such as p-toluenesulfonic acid, acetic acid, and the like.
  • Additional excipients which are contemplated for use in the practice of the present invention are those available to those of ordinary skill in the art, for example, those found in the United States Pharmacopeia Vol. XXII and National Formulary Vol. XVII, U.S. Pharmacopeia Convention, Inc., Rockville, Md. (1989), the relevant contents of which is incorporated herein by reference.
  • composition of the invention is a pharmaceutical composition.
  • Such compositions comprise a prophylactically or therapeutically effective amount of one or more prophylactic or therapeutic agents, including one or more of an HDAC inhibitor in combination with a therapeutically effective amount of one or more of an IGF-IR inhibitor, and/or an mTOR inhibitor, and optionally, an anti-androgen agent, and a pharmaceutically acceptable carrier.
  • the term "pharmaceutically acceptable” means approved by a regulatory agency of the Federal or a state government or listed in the U.S. Pharmacopeia or other generally recognized pharmacopeia for use in animals, and more particularly in humans.
  • carrier refers to a diluent, adjuvant (e.g., Freund's adjuvant (complete and incomplete)), excipient, or vehicle with which the therapeutic is administered.
  • Such pharmaceutical carriers can be sterile liquids, such as water and oils, including those of petroleum, animal, vegetable or synthetic origin, such as peanut oil, soybean oil, mineral oil, sesame oil and the like. Water is a preferred carrier when the pharmaceutical composition is administered intravenously.
  • Saline solutions and aqueous dextrose and glycerol solutions can also be employed as liquid carriers, particularly for injectable solutions.
  • suitable pharmaceutical excipients include starch, glucose, lactose, sucrose, gelatin, malt, rice, flour, chalk, silica gel, sodium stearate, glycerol monostearate, talc, sodium chloride, dried skim milk, glycerol, propylene, glycol, water, ethanol and the like.
  • the composition if desired, can also contain minor amounts of wetting or emulsifying agents, or pH buffering agents. These compositions can take the form of solutions, suspensions, emulsion, tablets, pills, capsules, powders, sustained-release formulations and the like.
  • Oral formulation can include standard carriers such as pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharine, cellulose, magnesium carbonate, etc. Examples of suitable pharmaceutical carriers are described in "Remington's Pharmaceutical Sciences” by E. W. Martin.
  • Such compositions will contain a prophylactically or therapeutically effective amount of a prophylactic or therapeutic agent preferably in purified form, together with a suitable amount of carrier so as to provide the form for proper administration to the patient.
  • the formulation should suit the mode of administration.
  • the pharmaceutical compositions are sterile and in suitable form for administration to a subject, preferably an animal subject, more preferably a mammalian subject, and most preferably a human subject.
  • compositions of the invention may be desirable to administer locally to the area in need of treatment; this may be achieved by, for example, and not by way of limitation, local infusion, by injection, or by means of an implant, said implant being of a porous, non-porous, or gelatinous material, including membranes, such as sialastic membranes, or fibers.
  • an implant being of a porous, non-porous, or gelatinous material, including membranes, such as sialastic membranes, or fibers.
  • care must be taken to use materials to which the prophylactic or therapeutic agents do not absorb.
  • the composition can be delivered in a vesicle, in particular a liposome (see Langer, Science 249:1527-1533 (1990); Treat et al., in Liposomes in the Therapy of Infectious Disease and Cancer, Lopez-Berestein and Fidler (eds.), Liss, New York, pp. 353-365 (1989); Lopez-Berestein, pp. 317-327; see generally above.).
  • a liposome see Langer, Science 249:1527-1533 (1990); Treat et al., in Liposomes in the Therapy of Infectious Disease and Cancer, Lopez-Berestein and Fidler (eds.), Liss, New York, pp. 353-365 (1989); Lopez-Berestein, pp. 317-327; see generally above.).
  • the composition can be delivered in a controlled release or sustained release system.
  • a pump may be used to achieve controlled or sustained release (see Langer, supra; Sefton, 1987, CRC Crit. Ref. Biomed. Eng. 14:20; Buchwald et al., 1980, Surgery 88:507; Saudek et al., 1989, N. Engl. J. Med. 321 :574).
  • polymeric materials can be used to achieve controlled or sustained release of the antibodies of the invention or fragments thereof (see e.g., Medical Applications of Controlled Release, Langer and Wise (eds.), CRC Pres., Boca Raton, FIa.
  • polymers used in sustained release formulations include, but are not limited to, poly(2-hydroxy ethyl methacrylate), poly(methyl methacrylate), poly(acrylic acid), poly(ethylene-co-vinyl acetate), poly(methacrylic acid), polyglycolides (PLG), polyanhydrides, poly(N-vinyl pyrrolidone), poly(vinyl alcohol), polyacrylamide, poly(ethylene glycol), polylactides (PLA), poly(lactide-co-glycolides) (PLGA), and polyorthoesters.
  • the polymer used in a sustained release formulation is inert, free of leachable impurities, stable on storage, sterile, and biodegradable.
  • a controlled or sustained release system can be placed in proximity of the therapeutic target, i.e., the lungs, thus requiring only a fraction of the systemic dose (see, e.g., Goodson, in Medical Applications of Controlled Release, supra, vol. 2, pp. 115-138 (1984)).
  • Controlled release systems are discussed in the review by Langer (1990, Science 249:1527-1533). Any technique known to one of skill in the art can be used to produce sustained release formulations comprising one or more antibodies of the invention or fragments thereof. See, e.g., U.S. Pat. No. 4,526,938, International publication No. WO 91/05548, International publication No.
  • the nucleic acid can be administered in vivo to promote expression of its encoded prophylactic or therapeutic agents, by constructing it as part of an appropriate nucleic acid expression vector and administering it so that it becomes intracellular, e.g., by use of a retroviral vector (see U.S. Pat. No.
  • a nucleic acid can be introduced intracellularly and incorporated within host cell DNA for expression by homologous recombination.
  • a pharmaceutical composition of the invention is formulated to be compatible with its intended route of administration.
  • suitable routes of administration include, but are not limited to, parenteral (e.g., intravenous, intramuscular, intradermal, intra-tumoral, intra-synovial, and subcutaneous), oral (e.g., inhalation), intranasal, transdermal (topical), transmucosal, intra-tumoral, intra-synovial, vaginal, and rectal administration.
  • the composition is formulated in accordance with routine procedures as a pharmaceutical composition adapted for intravenous, subcutaneous, intramuscular, oral, intra-tummoral, intra synnovial, intranasal or topical administration to human beings.
  • compositions for intravenous administration are solutions in sterile isotonic aqueous buffer.
  • the composition may also include a solubilizing agent and a local anesthetic such as lignocaine to ease pain at the site of the injection.
  • compositions of the invention are to be administered topically, the compositions can be formulated in the form of, e.g., a toothpaste, ointment, cream, transdermal patch, lotion, gel, oral gel, shampoo, spray, aerosol, solution, emulsion, or other form well-known to one of skill in the art. See, e.g., Remington's Pharmaceutical Sciences and Introduction to Pharmaceutical Dosage Forms, 4.sup.th ed., Lea & Febiger, Philadelphia, Pa. (1985).
  • viscous to semi-solid or solid forms comprising a carrier or one or more excipients compatible with topical application and having a dynamic viscosity preferably greater than water are typically employed.
  • suitable formulations include, without limitation, solutions, suspensions, emulsions, creams, ointments, powders, liniments, salves, and the like, which are, if desired, sterilized or mixed with auxiliary agents (e.g., preservatives, stabilizers, wetting agents, buffers, or salts) for influencing various properties, such as, for example, osmotic pressure.
  • suitable topical dosage forms include sprayable aerosol preparations wherein the active ingredient, preferably in combination with a solid or liquid inert carrier, is packaged in a mixture with a pressurized volatile (e.g., a gaseous propellant, such as freon), or in a squeeze bottle.
  • a pressurized volatile e.g., a gaseous propellant, such as freon
  • Moisturizers or humectants can also be added to pharmaceutical compositions and dosage forms if desired. Examples of such additional ingredients are well-known in the art.
  • compositions of the invention are to be administered intranasally, the compositions can be formulated in an aerosol form, spray, mist or in the form of drops.
  • prophylactic or therapeutic agents for use according to the present invention can be conveniently delivered in the form of an aerosol spray presentation from pressurized packs or a nebuliser, with the use of a suitable propellant, e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas.
  • a suitable propellant e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas.
  • the dosage unit may be determined by providing a valve to deliver a metered amount.
  • Capsules and cartridges of, e.g., gelatin for use in an inhaler or insufflator may be formulated containing
  • compositions of the invention are to be administered orally, the compositions can be formulated orally in the form of, e.g., gum, tablets, capsules, cachets, gelcaps, solutions, suspensions and the like. Tablets or capsules can be prepared by conventional means with pharmaceutically acceptable excipients such as binding agents (e.g., pregelatinised maize starch, polyvinylpyrrolidone or hydroxypropyl methylcellulose); fillers (e.g., lactose, microcrystalline cellulose or calcium hydrogen phosphate); lubricants (e.g., magnesium stearate, talc or silica); disintegrants (e.g., potato starch or sodium starch glycolate); or wetting agents (e.g., sodium lauryl sulphate).
  • binding agents e.g., pregelatinised maize starch, polyvinylpyrrolidone or hydroxypropyl methylcellulose
  • fillers e.g., lactose, microcrystalline cellulose
  • Liquid preparations for oral administration may take the form of, for example, solutions, syrups or suspensions, or they may be presented as a dry product for constitution with water or other suitable vehicle before use.
  • Such liquid preparations may be prepared by conventional means with pharmaceutically acceptable additives such as suspending agents (e.g., sorbitol syrup, cellulose derivatives or hydrogenated edible fats); emulsifying agents (e.g., lecithin or acacia); non-aqueous vehicles (e.g., almond oil, oily esters, ethyl alcohol or fractionated vegetable oils); and preservatives (e.g., methyl or propyl-p-hydroxybenzoates or sorbic acid).
  • the preparations may also contain buffer salts, flavoring, coloring and sweetening agents as appropriate.
  • Preparations for oral administration may be suitably formulated for slow release, controlled release or sustained release of a prophylactic or therapeutic agent(s).
  • compositions of the invention may be formulated for parenteral administration by injection, e.g., by bolus injection or continuous infusion.
  • Formulations for injection may be presented in unit dosage form, e.g., in ampoules or in multi-dose containers, with an added preservative.
  • the compositions may take such forms as suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents.
  • the active ingredient may be in powder form for constitution with a suitable vehicle, e.g., sterile pyrogen-free water, before use.
  • compositions of the invention may also be formulated in rectal compositions such as suppositories or retention enemas, e.g., containing conventional suppository bases such as cocoa butter or other glycerides.
  • compositions of the invention may also be formulated as a depot preparation. Such long acting formulations may be administered by implantation (for example subcutaneously or intramuscularly) or by intramuscular injection.
  • the compositions may be formulated with suitable polymeric or hydrophobic materials (for example as an emulsion in an acceptable oil) or ion exchange resins, or as sparingly soluble derivatives, for example, as a sparingly soluble salt.
  • compositions of the invention can be formulated as neutral or salt forms.
  • Pharmaceutically acceptable salts include those formed with anions such as those derived from hydrochloric, phosphoric, acetic, oxalic, tartaric acids, etc., and those formed with cations such as those derived from sodium, potassium, ammonium, calcium, ferric hydroxides, isopropylamine, triethylamine, 2-ethylamino ethanol, histidine, procaine, etc.
  • compositions of the invention are supplied either separately or mixed together in unit dosage form, for example, as a dry lyophilized powder or water free concentrate in a hermetically sealed container such as an ampoule or sachette indicating the quantity of active agent.
  • a hermetically sealed container such as an ampoule or sachette indicating the quantity of active agent.
  • the composition is to be administered by infusion, it can be dispensed with an infusion bottle containing sterile pharmaceutical grade water or saline.
  • an ampoule of sterile water for injection or saline can be provided so that the ingredients may be mixed prior to administration.
  • the invention provides a pharmaceutical pack or kit comprising one or more containers filled with individual components (in pharmaceutical formulations) of the combination therapies described herein; for example, contained filled with one or more prophylactic or therapeutic agents, including one or more of an HDAC inhibitor in combination with a therapeutically effective amount of one or more of an IGF-IR inhibitor, and/or an mTOR inhibitor, and optionally, an anti-androgen agent, as well as other active ingredients, such as chemotherapy agents.
  • the pharmaceutical pack or kit may further comprises one or more other prophylactic or therapeutic agents useful for the treatment of a disease or disorder.
  • the invention also provides a pharmaceutical pack or kit comprising one or more containers filled with one or more of the ingredients of the pharmaceutical compositions of the invention.
  • Optionally associated with such container(s) can be a notice in the form prescribed by a governmental agency regulating the manufacture, use or sale of pharmaceuticals or biological products, which notice reflects approval by the agency of manufacture, use or sale for human administration.
  • the present invention also encompasses a finished packaged and labeled pharmaceutical product.
  • This article of manufacture includes the appropriate unit dosage form in an appropriate vessel or container such as a glass vial or other container that is hermetically sealed.
  • the active ingredient is sterile and suitable for administration as a particulate free solution.
  • the unit dosage form may be a solid suitable for oral, transdermal, intratumoral, intra-synovial, topical or mucosal delivery.
  • the unit dosage form is suitable for intravenous, intramuscular, intratumoral, intra-synovial, or subcutaneous delivery.
  • the invention encompasses solutions, preferably sterile, suitable for each delivery route.
  • the packaging material and container are designed to protect the stability of the product during storage and shipment.
  • the products of the invention include instructions for use or other informational material that advise the physician, technician or patient on how to appropriately prevent or treat the disease or disorder in question.
  • the article of manufacture includes instruction means indicating or suggesting a dosing regimen including, but not limited to, actual doses, monitoring procedures (such as methods for monitoring mean absolute lymphocyte counts, tumor cell counts, calcium concentration, and tumor size) and other monitoring information.
  • the invention provides an article of manufacture comprising packaging material, such as a box, bottle, tube, vial, container, sprayer, insufflator, intravenous (i.v.) bag, envelope and the like; and at least one unit dosage form of a pharmaceutical agent contained within said packaging material.
  • packaging material such as a box, bottle, tube, vial, container, sprayer, insufflator, intravenous (i.v.) bag, envelope and the like
  • a unit dosage form of a pharmaceutical agent contained within said packaging material such as a box, bottle, tube, vial, container, sprayer, insufflator, intravenous (i.v.) bag, envelope and the like.
  • an article of manufacture comprises packaging material and a pharmaceutical agent and instructions contained within said packaging material, wherein said pharmaceutical agent comprises at least one or more prophylactic or therapeutic agents, including one or more of an HDAC inhibitor in combination with a therapeutically effective amount of one or more of an IGF-IR inhibitor, and/or an mTOR inhibitor, and optionally, an anti-androgen agent and a pharmaceutically acceptable carrier, and said instructions indicate a dosing regimen for preventing, treating or managing a subject with cancer.
  • the compounds utilized in the pharmaceutical method of the invention are administered at the initial dosage of about 0.001 mg/kg to about 1000 mg/kg daily.
  • the dosages may be varied depending upon the requirements of the patient, the severity of the condition being treated, and the compound being employed. For example, dosages can be empirically determined considering the type and stage of cancer diagnosed in a particular patient.
  • the dose administered to a patient should be sufficient to effect a beneficial therapeutic response in the patient over time.
  • the size of the dose also will be determined by the existence, nature, and extent of any adverse side-effects that accompany the administration of a particular compound in a particular patient. Determination of the proper dosage for a particular situation is within the skill of the practitioner. Generally, treatment is initiated with smaller dosages which are less than the optimum dose of the compound. Thereafter, the dosage is increased by small increments until the optimum effect under circumstances is reached. For convenience, the total daily dosage may be divided and administered in portions during the day, if desired. Doses can be given daily, or on alternate days, as determined by the treating physician.
  • Toxicity and efficacy of the prophylactic and/or therapeutic treatments and protocols of the instant invention can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., for determining the LD50 (the dose lethal to 50% of the population) and the ED50 (the dose therapeutically effective in 50% of the population).
  • the dose ratio between toxic and therapeutic effects is the therapeutic index and it can be expressed as the ratio LD50/ED50.
  • Prophylactic and/or therapeutic agents that exhibit large therapeutic indices are preferred. While prophylactic and/or therapeutic agents that exhibit toxic side effects may be used, care should be taken to design a delivery system that targets such agents to the site of affected tissue in order to minimize potential damage to uninfected cells and, thereby, reduce side effects.
  • the data obtained from the cell culture assays and animal studies can be used in formulating a range of dosage of the prophylactic and/or therapeutic agents for use in humans.
  • the dosage of such agents lies preferably within a range of circulating concentrations that include the ED 50 with little or no toxicity.
  • the dosage may vary within this range depending upon the dosage form employed and the route of administration utilized.
  • the therapeutically effective dose can be estimated initially from cell culture assays.
  • a dose may be formulated in animal models to achieve a circulating plasma concentration range that includes the IC50 (i.e., the concentration of the test compound that achieves a half- maximal inhibition of symptoms) as determined in cell culture. Such information can be used to more accurately determine useful doses in humans.
  • Levels in plasma may be measured, for example, by high performance liquid chromatography.
  • Also encompassed by the invention to determine therapeutic or prophylactic utility are any relevant cancer, and more specifically, prostate cancer animal models.
  • TRAMP model that utilizes the minimal rat probasin promoter to express the SV40 early genes (T and t antigens; Tag) as well as a number of transgenic lines using the long probasin promoter to express large T antigen, collectively termed the 'LADY' model.
  • the Cryptdin-2-T and Gg-SV40 T models also develop progressive prostate cancer and may be used in the invention. Further encompassed by the invention, pending safety and efficacy, are clinical trials to assess the combinations of the present invention.
  • the HDAC inhibitor TSA combines effectively to reduce proliferation of IGF- IR inhibitors (EGCG and PPP) and the mTOR inhibitor rapamycin in prostate cancer cells.
  • IGF- IR inhibitors EGCG and PPP
  • rapamycin the mTOR inhibitor rapamycin in prostate cancer cells.
  • IGF- IR inhibitors EGCG and PPP
  • rapamycin the mTOR inhibitor rapamycin in prostate cancer cells.
  • TSA also combines effectively with PPP in androgen-sensitive LNCaP cells.
  • the HDAC inhibitor VPA combines effectively to reduce proliferation of IGF-IR inhibitors (EGCG and PPP) and the mTOR inhibitor rapamycin in prostate cancer cells in a cell type specific manner.
  • IGF-IR inhibitors EGCG and PPP
  • rapamycin in androgen-sensitive LNCaP cells, the combination of VPA and EGCG is more efficacious than either drug used alone.
  • VPA also combines effectively with rapamycin to inhibit LNCaP cell proliferation better than either drug used alone.
  • in androgen-insensitive DU 145 cells SFN and rapamycin combine effectively to inhibit cell proliferation, and a triple combination of SFN with an IGF-IR inhibitor (PPP or EGCG) and rapamycin is more efficacious than any one of these drugs alone or any two drugs in combination.
  • VPA combines effectively with bicalutamide to reduce proliferation in androgen-sensitive prostate cells.
  • a dose of 5OnM bicalutamide was chosen as an effective dose to inhibit androstendiol-stimulated cell proliferation.
  • VPA inhibited cell proliferation by itself and also increased the efficacy of bicalutamide.
  • the anti-androgen, bicalutamide combines effectively with VPA and EGCG to inhibit androgen-sensitive prostate cell proliferation.
  • a dose of 1 ⁇ M bicalutamide was chosen as an effective dose to inhibit androstenediol-stimulated cell proliferation (see FIG. 4A).
  • the triple combination of VPA, EGCG and bicalutamide is more effective at inhibiting androgen-sensitive prostate cell proliferation than any one of these drugs or double combination of the drugs.
  • Bicalutamide and VPA combine effectively to inhibit androgen-sensitive prostate cell proliferation, as do bicalutamide and EGCG (see FIG. 4B).
  • HDAC inhibitors SFN, TSA, and VPA alone or in combination with rapamycin, further in combination with either the IGF-IR inhibitor, EGCG or PPP on cell proliferation of the human prostate cancer cell line, LNCaP after being in culture for 6 days. Proliferation was measured using a DNA-binding fluorescent dye. Cells were treated with the following doses of inhibitors: 500 ⁇ M VPA, 2nM TSA, 1 ⁇ M SFN, 7 ⁇ M EGCG, 3OnM PPP, 2nM rapamycin (see FIG. 1).

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

The present application demonstrates that HDAC inhibitors can be used in combination with IGF-1R inhibitors and mTOR inhibitors to treat prostate cancer. Combinations of the compounds, with or without HDAC inhibitors, and with or without androgen ablation therapy, can also be used, if indicated. The invention therefore provides methods of treatment and pharmaceutical compositions.

Description

COMBINATION THERAPY FOR PROSTATE CANCER
BACKGROUND OF THE INVENTION
[0001] Prostate cancer is frequently detected at an early stage using the prostate specific antigen (PSA) test and subsequent biopsy. It is treated most often either with surgical removal of the prostate or by destruction of the prostate with radiation or other means. Men with early stage prostate cancer also elect "watchful waiting" in which surgery is delayed or suspended until the cancer worsens. In addition there are substantial numbers of men with pre-cancerous conditions of the prostate such as prostatic intraepithelial neoplasia (PIN) on watchful waiting because these conditions frequently progress to cancer.
[0002] Currently there is no treatment that delays or abolishes the progression from PIN to prostate cancer, nor is there a treatment to delay or abolish the progression from early to more advanced prostate cancer in men on watchful waiting.
[0003] After surgery, prostate cancer can recur, and is often detected by an increase in prostate specific antigen (PSA) after the initial post-surgical decline. Recurrent prostate cancer is often treated with androgen ablation therapy, an effective treatment for this stage of the disease where growth of tumor cells is dependent on androgen stimulation. Unfortunately, the response to androgen ablation is limited as the tumor progresses to androgen independence via mutations that make proliferation of the tumor cells independent of the action of the androgen receptor.
[0004] Currently there are few options for treatment once androgen ablation therapy fails. Chemotherapy is sometimes used but the efficacy is not acceptable. In addition, there is no treatment to delay the progression of recurrent prostate cancer (as measured by PSA) and thereby delay the time at which androgen ablation therapy must begin.
[0005] Histone deacetylase (HDAC) inhibitors are a structurally diverse group of pharmacological agents that inhibit proliferation, induce differentiation and/or apoptosis in a wide range of cancer cells and hold much promise as anti-neoplastic agents (Villar- Garea, A. and M. Esteller, Int J Cancer, 2004. 112(2): p. 171-8; Marks, P., et al, Nat Rev Cancer, 2001. 1(3): p. 194-202). Hyperacetylation is associated with a transcriptionally permissive environment and HDAC inhibitors, although they affect only a small number of target genes, activate genes involved in cell cycle arrest, apoptosis and differentiation (Glaser, K.B., et al, MoI Cancer Ther, 2003. 2(2): p. 151-63; Richon, V.M., et al, Proc Natl Acad Sci USA, 2000. 97(18): p. 10014-9; Munster, P.N., et al., Cancer Res., 2001. 61(23): p. 8492-7). Furthermore, HDAC inhibitors increase the efficiency of several anticancer drugs that target the DNA (Kim, M.S., et al., Cancer Res, 2003. 63(21): p. 7291-300; Castro-Galache, M.D., et al., Int J Cancer, 2003. 104(5): p. 579-86). A variety of small molecule HDAC inhibitors are currently in preclinical development.
[0006] Given the unacceptable treatments currently available, there is a compelling need to develop new therapeutic strategies for the treatment and prevention of cancer, and in particular, for the treatment, prevention of, and prevention of progression of prostate cancer.
SUMMARY OF THE INVENTION
[0007] The present inventors have investigated the ability of cocktails of targeted anticancer agents, including HDAC inhibitors, to inhibit the proliferation of cell lines derived from various stages of prostate cancer; androgen dependent, androgen independent (albeit AR positive), and completely androgen independent (AR negative). The inventors have discovered that, surprisingly, certain HDAC inhibitors combine effectively with other therapies for prostate cancer cells at particular stages, and that the combinations do not reduce the activity of any single component, and that these combinations are more effective than either of the components alone. For example, the inventors have investigated the effects of various combinations of HDAC inhibitors including VPA, sulforaphane (SFN), and trichostatin (TSA) in combination with the mTOR inhibitor rapamycin, the IGF-IR inhibitor picropodophyllin (PPP), and the green tea polyphenol, epigallocatechin gallate (EGCG), also an IGF-IR inhibitor, as well as androgen ablation therapy, such as bicalutamide. The findings indicate that the HDAC inhibitors combine effectively with the inhibitory actions of the other anti-cancer compounds.
[0008] In particular, the present inventors have found that for certain stages of prostate cancer, certain combinations of HDAC inhibitors and one or more other compounds, including, but not limited to IGF-IR inhibitors, mTOR inhibitors and androgen ablation therapy combine effectively with each other and are superior to other combinations of compounds for the treatment and prevention of prostate cancer, and for preventing the progression of prostate cancer. The data provided herein show that certain agents that inhibit prostate cancer cell proliferation on their own, combine effectively for prostate cancer cells from certain stages of cancer but, surprisingly, not for others (for example, in androgen-dependent prostate cancer cells, such as LNCaP cells versus androgen independent prostate cancer cells, such as DU145 cells). The present inventors have found that the combination of VPA and EGCG (both of which inhibit prostate cancer cell proliferation as single agents) was effective on earlier stage (androgen- dependent) prostate cancer cells, but not on later stage (androgen-independent) prostate cancer cells. On the other hand, however, the combination of the HDAC inhibitor, SFN and the mTOR inhibitor, rapamycin, is effective against later stage (androgen- independent) prostate cancer cells. These results are surprising and were unpredictable prior to the present invention. Accordingly, the present invention provides methods of treating and preventing prostate cancer at various stages, including androgen dependent, androgen independent but androgen receptor (AR) positive, and completely androgen independent (AR negative) prostate cancer using the combinations that are effective for the particular stage of cancer, as well as pharmaceutical compositions comprising the compounds used in the combination therapies.
[0009] The present invention encompasses methods of treatment for (including management of, amelioration of symptoms of, and preventing the progression of) prostate cancer, using certain combination therapies, as well as the pharmaceutical compositions comprising these combination therapies. The invention is based, in part, on the recognition that certain combinations of compounds combine effectively with each other, and are superior to other combinations of compounds, as well as improving the tolerance of, and/or reducing the side effects caused by at least one of the compounds in the combination. Subjects are mammalian, and preferably are human, and more preferably are human males.
[0010] In one aspect, the present invention encompasses methods of treating and methods of preventing prostate cancer comprising administering to a subject suffering therefrom a therapeutically effective amount of an HDAC inhibitor in combination with a therapeutically effective amount of one or more of an IGF-I receptor inhibitor, and/or an mTOR inhibitor, and optionally, androgen ablation therapy. In some embodiments, various combinations of these compounds, or a single one of the compounds, is administered in combination with androgen ablation therapy. In one embodiment, one or more of the compounds are administered in combination with an anti-androgen, such as bicalutamide.
[0011] The present invention also encompasses methods and compositions for preventing prostate cancer, particularly in subjects who are at risk for prostate cancer that is greater than the average risk for prostate cancer. Risk factors considered in preventing prostate cancer in subjects include, but are not limited to those diagnosed with prostatic intraepithelial neoplasia (PIN), prostate-specific antigen (PSA) levels, family history of prostate cancer (relatives with prostate cancer), age, age at which relatives were diagnosed, circulating hormone levels, genetic markers for prostate cancer such as PCA3 and 8q24, diet, obesity, race, ethnicity, nationality, whether the individual has had a vasectomy, and presence of prostatitis. The present invention also provides methods and compositions for preventing the progression of prostate cancer to a later stage for those who already have prostate cancer or precancerous indicators, as well as preventing the recurrence of prostate cancer for those in remission from prostate cancer.
[0012] In some embodiments, the invention contemplates methods of preventing the progression of prostatic intraepithelial neoplasia (PIN) and methods of preventing the progression of PIN to prostate cancer. In some preferred embodiments, the invention encompasses treating or preventing androgen (AR)-dependent prostate cancer, in some preferred embodiments, the invention encompasses treating or preventing androgen independent (albeit AR positive) prostate cancer, and in yet other preferred embodiments, the invention encompasses treating or preventing completely androgen independent (AR negative) prostate cancer.
[0013] In yet another embodiment, the present invention encompasses treating PIN, including low grade (PIN I) and high grade (PIN II and PIN III) lesions.
[0014] In another aspect, the present invention encompasses methods of treating and methods of preventing prostate cancer comprising administering to a subject suffering therefrom a therapeutically effective amount of an HDAC inhibitor in combination with a therapeutically effective amount of one or more of an IGF-I receptor inhibitor, or an mTOR inhibitor, in combination with androgen ablation therapy, in individuals with androgen-dependent prostate cancer. In one embodiment, one or more of the compounds are administered in combination with bicalutamide.
[0015] The present invention also provides pharmaceutical compositions comprising or consisting essentially of the combinations of compounds described herein, as well as kits comprising the combinations.
[0016] The HDAC inhibitors encompassed by the present invention can be any known to those of skill in the art, including those described in Minucci et al, Nature 6:38- 51 (2006). According to the invention, in some embodiments, the HDAC inhibitor is VPA. In other embodiments, the HDAC inhibitor may be SFN or TSA. In yet other embodiments, the HDAC inhibitor may be SAHA or any other HDAC inhibitor known by the skilled practitioner to be effective.
[0017] In one embodiment, the patient is treated with a combination of an HDAC inhibitor, and one or more of an IGF-IR inhibitor, and/or one or more of an mTOR inhibitor. Optionally, the combination also comprises androgen ablation therapy. In another embodiment, the patient is treated with a combination of an HDAC inhibitor and an IGF-IR inhibitor. In yet another embodiment, the patient is treated with a combination of an HDAC inhibitor and an mTOR inhibitor. Optionally, the combination also comprises androgen ablation therapy.
[0018] In another embodiment, a patient with AR-responsive, early-stage prostate cancer is treated with a combination of VPA and EGCG. In another embodiment, a patient with AR-responsive, early-stage prostate cancer is treated with VPA, EGCG, and rapamycin. In one embodiment, one or more of the compounds are administered in combination with the bicalutamide. In other embodiments, these combinations are used to prevent the progression from pre-cancerous stages to prostate cancer in patients.
[0019] In another embodiment, a patient with AR-independent prostate cancer, and particularly AR-independent prostate cancer which is still AR-positive, is treated with a combination of SFN and EGCG. In another embodiment, a patient with AR-independent prostate cancer, and in particular, AR-independent prostate cancer which is still AR- positive, is treated with a combination of SFN and PPP.
[0020] In another embodiment, a patient with AR-independent prostate cancer, either still AR positive or completely AR negative, is treated with a combination of SFN and rapamycin. [0021] In yet another embodiment, a patient is treated with a combination of an HDAC inhibitor, an IGF-IR inhibitor, and an mTOR inhibitor. In one embodiment, the patient is treated with rapamycin, PPP, and SFN.
[0022] In yet another embodiment, a patient with AR-responsive prostate cancer is treated with bicalutamide and VPA.
[0023] In another embodiment, a patient with AR-responsive prostate cancer is treated with bicalutamide, VPA, and EGCG.
[0024] In other embodiments, methods of preventing progression from PIN to prostate cancer, methods of preventing progression from low to higher grade prostate cancer, and methods of preventing recurrence of prostate cancer comprise administering a combination of an HDAC inhibitor, and one or more of an IGF-IR inhibitor, and/or one or more of an mTOR inhibitor. Optionally, the combination also comprises androgen ablation therapy.
[0025] In yet other embodiments, methods of slowing progression of naϊve (not yet treated with androgen ablation therapy) recurrent prostate cancer comprise administering a combination of an HDAC inhibitor, and one or more of an IGF-IR inhibitor, and/or one or more of an mTOR inhibitor. Optionally, the combination also comprises androgen ablation therapy.
[0026] In another embodiment, methods of preventing resistance to androgen ablation therapy comprise administering a combination of an HDAC inhibitor, and one or more of an IGF-IR inhibitor, and/or one or more of an mTOR inhibitor, in addition to androgen ablation therapy.
[0027] In one preferred embodiment, the dose of valproic acid is between about 2g/day and about 3g/day.
[0028] In one embodiment, the daily dose of valproic acid is from about 15/mg/kg to about 60 mg/kg.
[0029] In one embodiment, the daily dose of valproic acid is sufficient to achieve about 300 to about 867 micromolar in patient serum. In another embodiment, the daily dose of valproic acid is sufficient to achieve about 300 to about 1000 micromolar in patient serum, and in another embodiment, the daily dose of valproic acid is sufficient to achieve about 500 to about 1000 micromolar in patient serum. [0030] In some embodiments, the dose of SAHA is from about 200 mg/day to about 600 mg/day. In another embodiment, the dose of SAHA is about 400 mg/day.
[0031] In some embodiments, compounds are administered in combination, with ratios of those compounds which preserve the recommended daily doses of the compounds. In some embodiments, compounds are administered in combination, with ratios of those compounds which preserve the ranges of doses as described herein.
[0032] The present invention further contemplates that the combination therapies as described herein can also reduce or eliminate other side effects of treatment, at least in part because lower doses of compounds can be used in treatment or prevention protocols.
[0033] In certain of the embodiments, the IGF-IR inhibitor can be picropodophyllin (see, e.g., Girnita, A. et al, Cancer Res., 2004. 64(1): 236-242) or the green tea polyphenol, EGCG (see, e.g., Shimizu, M. et al, Biochem. Biophys. Res. Commun., 2005. 334(3): 947-953; Li, M. et al., Cancer Epidemiol. Biomarkers Prev., 2007. 16(3): 598- 605. The mTOR inhibitor may be rapamycin or rapamycin derivatives (see, e.g., Johnston, S.R., Clin. Cancer Res., 2006. 12(3 Pt. 2): 1061-1069s). The skilled practitioner will be able to use a variety of IGF-IR and mTOR inhibitors in the invention, to provide therapeutically effective combinations with various HDAC inhibitors.
[0034] In one embodiment, the dose of EGCG is from about 800 mg/day to about 1200 mg/day.
[0035] In another embodiment, the dose of EGCG is from about 300 mg/day to about 800 mg/day.
[0036] In one embodiment, the dose of rapamycin is from about 0.125 mg/day to about 1 mg/day.
[0037] In one embodiment, the combination comprises a ratio of VPA: EGCG from about 2 to about 3 parts VPA to about 1 part EGCG.
[0038] In some embodiments, an additional "active ingredient" that can be used in combination with combination therapies described above. In different embodiments, the additional active ingredient a other chemotherapeutic agent, biologic, radiation therapy, or other agents and procedures useful in the treatment of cancer, including androgen ablation therapy. [0039] Various drug administration protocols are contemplated by the invention. In some embodiments, the HDAC inhibitor is administered on a daily basis, while the IGF- IR inhibitor and/or mTOR inhibitor is administered every other day. In other embodiments, the IGF-IR inhibitor and/or mTOR inhibitor is administered on a daily basis and the HDAC inhibitor is administered every other day. The invention also contemplates administering the HDAC inhibitor and the -IR inhibitor and/or mTOR inhibitor concurrently.
[0040] It is within the scope of the invention to treat prostate cancer that is androgen- dependent, androgen-independent but AR positive, and androgen-independent. The invention also contemplates treating subjects with prostate cancer for whom previous therapy has failed, or for whom the cancer is recurring. In some embodiments, the invention contemplates treating subjects who are genetically predisposed to prostate cancer or otherwise at increased risk. The invention also encompasses methods of treating subjects to prevent progression of prostate cancer, and in some embodiments, the invention encompasses treating or preventing prostate cancer in patients with precancerous growths or benign tumors. It is within the scope of the invention to treat subjects that are in remission from prostate cancer, and to treat subjects with prostate cancer that have previously undergone treatment.
BRIEF DESCRIPTION OF THE DRAWINGS
[0041] FIGS. 1A-1C: The effect of the HDAC inhibitors, SFN, TSA, and VPA alone or in combination with rapamycin, further in combination with either the IGF-IR inhibitor, EGCG or PPP on cell proliferation of the human prostate cancer cell line, LNCaP after being in culture for 6 days. Cells were treated as indicated with the following doses of inhibitors: 500μM VPA, 2nM TSA, 1 μM SFN, 7μM EGCG, 3OnM PPP, 2nM rapamycin. Asterisks denote that a combination of drugs produces a more efficacious response than any of the drugs used alone.
[0042] FIGS. 2A-2C: The effect of the HDAC inhibitors, SFN, TSA, and VPA alone or in combination with rapamycin, further in combination with either the IGF-IR inhibitor, EGCG or PPP on cell proliferation of the human prostate cancer cell line, DU145 after being in culture for 6 days. Cells were treated as indicated with the following doses of inhibitors: 500μM VPA, 2nM TSA, 1 μM SFN, 7μM EGCG, 3OnM PPP, 2nM rapamycin. Asterisks denote that a combination of drugs produces a more efficacious response than any of the drugs used alone.
[0043] FIGS. 3A-3C: The effect of the HDAC inhibitors, SFN, TSA, and VPA alone or in combination with rapamycin, further in combination with either the IGF-IR inhibitor, EGCG or PPP on cell proliferation of the human prostate cancer cell line, PC3 after being in culture for 6 days. Cells were treated as indicated with the following doses of inhibitors: 500μM VPA, 2nM TSA, 1 μM SFN, 7μM EGCG, 3OnM PPP, 2nM rapamycin.
[0044] FIGS. 4A-4B: A) Dose response of the anti-androgen, bicalutamide in androgen-sensitive prostate cells. LNCaP cells were treated with the indicated doses of bicalutamide in the presence of 5OnM androstenediol. B) The effects of bicalutamide, the HDAC inhibitor, VPA, and the IGF-IR inhibitor EGCG, alone and in double or triple combination on cell proliferation of the human prostate cancer cell line, LNCaP after being in culture for 6 days.
DETAILED DESCRIPTION OF THE INVENTION
[0045] The present inventors have found that certain combinations of HDAC inhibitors and other compounds, including, but not limited to IGF-IR inhibitors, and mTOR inhibitors combine effectively with each other for the treatment and prevention of prostate cancer, and for preventing the progression of prostate cancer, as well as for preventing the progression of prostatic intraepithelial neoplasia (PIN). The inventors have discovered that, surprisingly, for certain stages of prostate cancer, certain combinations of HDAC inhibitors combine effectively with other anti-cancer compounds, and that the combinations are more effective than any of the components alone. The present invention provides methods of treating and preventing prostate cancer, as well as pharmaceutical compositions comprising the compounds used in the combination therapies disclosed herein.
[0046] The invention also provides combinations of HDAC inhibitors, including but not limited to SFN, TSA, SAHA, and VPA, as well as inhibitors of the mammalian target of rapamycin (mTOR) protein such as rapamycin or derivatives thereof, and inhibitors of the insulin-like growth factor receptor (IGF-IR) signaling pathway such as picropodophyllin and EGCG. The combination of HDAC inhibitor, IGF-IR inhibitor, and mTOR inhibitor can also be used, as well as other combinations as described herein, including combinations with androgen ablation therapy, such as bicalutamide. Each of these combinations can further be used in combination with other therapies to treat prostate cancer, as described herein.
[0047] In some embodiments, the individual compounds in the combination therapies combine effectively with each other, and in other embodiments, the individual compounds in the combination therapies synergize with each other.
Definitions
[0048] As used herein, the term "cancer" refers to a disease involving cells that have the potential to metastasize to distal sites and exhibit phenotypic traits that differ from those of non-cancer cells. Cancer cells acquire a characteristic set of functional capabilities during their development, albeit through various mechanisms. Such capabilities include evading apoptosis, self-sufficiency in growth signals, insensitivity to anti-growth signals, tissue invasion/metastasis, limitless replicative potential, and sustained angiogenesis. The term "cancer cell" is meant to encompass both pre -malignant and malignant cancer cells.
[0049] The terms "histone deacetylase inhibitor" and "inhibitor of histone deacetylase" mean a compound which is capable of interacting with a histone deacetylase and inhibiting its enzymatic activity. "Inhibiting histone deacetylase enzymatic activity" means reducing the ability of a histone deacetylase to remove an acetyl group from a histone. (see, e.g., FIG. 8 and Minucci et al, Nature 6:38-51 (2006). In some preferred embodiments, such reduction of histone deacetylase activity is at least about 50%, more preferably at least about 75%, and still more preferably at least about 90%. In other preferred embodiments, histone deacetylase activity is reduced by at least 95% and more preferably by at least 99%. Assays for determining inhibition are described in Phiel, C. J., et al., J Biol Chem., 2001. 276(39): p. 36734-41 and Gottlicher, M., et al., Embo J., 2001. 20(24): p. 6969-78.
[0050] Preferably, such inhibition is specific, i.e., the histone deacetylase inhibitor reduces the ability of a histone deacetylase to remove an acetyl group from a histone at a concentration that is lower than the concentration of the inhibitor that is required to produce another, unrelated biological effect. Preferably, the concentration of the inhibitor required for histone deacetylase inhibitory activity is at least 2-fold lower, more preferably at least 5 -fold lower, even more preferably at least 10-fold lower, and most preferably at least 20-fold lower than the concentration required to produce an unrelated biological effect.
[0051] As used herein, the term "active ingredient" includes having a therapeutic or prophylactic effect on prostate cancer in the combinations. This does not include inactive ingredients such as pharmaceutical carriers, excipients, and the like.
[0052] "Mammalian target of rapamycin protein inhibitor" or "mTOR inhibitor" includes drugs such as rapamycin, temsirolimus, and everolimus that selectively inhibit the mammalian target of rapamycin (mTOR).
[0053] "IGF-I receptor inhibitor" refers to drugs such as picrophodophyllin and podophyllotoxin that selectively inhibit the IGF-I receptor.
[0054] "Androgen ablation therapy" refers to any type of therapy, including drug treatment, procedures, or surgery, which reduces the amount of androgen in an individual.
[0055] As used herein, the terms "prevent," "preventing" and "prevention" refer to the prevention of the recurrence, worsening, or spread of a disease in a subject resulting from the administration of a prophylactic or therapeutic agent.
[0056] The terms "overexpress," "overexpression" or "overexpressed" interchangeably refer to a protein or nucleic acid (RNA) that is translated or transcribed at a detectably greater level, usually in a cancer cell, in comparison to a normal cell. The term includes overexpression due to transcription, post transcriptional processing, translation, post-translational processing, cellular localization (e.g., organelle, cytoplasm, nucleus, cell surface), and RNA and protein stability, as compared to a normal cell. Overexpression can be detected using conventional techniques for detecting mRNA (i.e., RT-PCR, PCR, hybridization, microarray) or proteins (i.e., ELISA, immunohistochemical techniques). Overexpression can be 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or more in comparison to a normal cell. In certain instances, overexpression is 1-fold, 2-fold, 3 -fold, 4-fold or more higher levels of transcription or translation in comparison to a normal cell.
[0057] As used herein, the term "in combination" refers to the use of more than one prophylactic and/or therapeutic agents. The use of the term "in combination" does not restrict the order in which prophylactic and/or therapeutic agents are administered to a subject with cancer, especially prostate cancer. A first prophylactic or therapeutic agent can be administered prior to (e.g., 1 minute, 5 minutes, 15 minutes, 30 minutes, 45 minutes, 1 hour, 2 hours, 4 hours, 6 hours, 12 hours, 24 hours, 48 hours, 72 hours, 96 hours, 1 week, 2 weeks, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 8 weeks, or 12 weeks before), concomitantly with, or subsequent to (e.g., 1 minute, 5 minutes, 15 minutes, 30 minutes, 45 minutes, 1 hour, 2 hours, 4 hours, 6 hours, 12 hours, 24 hours, 48 hours, hours, 96 hours, 1 week, 2 weeks, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 8 weeks, or 12 weeks after) the administration of a second prophylactic or therapeutic agent to a subject which had, has, or is susceptible to cancer, especially prostate cancer. The prophylactic or therapeutic agents are administered to a subject in a sequence and within a time interval such that the agent of the invention can act together with the other agent to provide an increased benefit than if they were administered otherwise. Any additional prophylactic or therapeutic agent can be administered in any order with the other additional prophylactic or therapeutic agents.
[0058] As used herein, the term "combine effectively" refers to a combination of therapies (e.g., a combination of prophylactic or therapeutic agents) which is more effective than any single agent administered alone. Combining effectively may also refer to combinations of therapies that are not less effective than any single agent or even less effective than any single agent, but which also eliminate or reduce the adverse effects of one or more of the agents.
[0059] As used herein, the term "synergistic" refers to a combination of therapies (e.g., a combination of prophylactic or therapeutic agents) which is more effective than the additive effects of any two or more single agents. A synergistic effect of a combination of therapies permits the use of lower dosages of one or more of the therapies and/or less frequent administration of said therapies (e.g., agents) to a subject with a disease or disorder, in particular, cancer, or a condition or symptom associated therewith. The ability to utilize lower dosages of therapies and/or to administer said therapies less frequently reduces the toxicity associated with the administration of said therapies to a subject without reducing the efficacy of said therapies in the prevention, management, or treatment of a disease or disorder, in particular, cancer or a condition or symptom associated therewith. In addition, a synergistic effect can result in improved efficacy of therapies in the prevention, management, or treatment of a disease or disorder, in particular, cancer or a condition or symptom associated therewith. Finally, the synergistic effect of a combination of therapies may avoid or reduce adverse or unwanted side effects associated with the use of any single therapy. [0060] As used herein, the phrase "side effects" encompasses unwanted and adverse effects of a prophylactic or therapeutic agent. Adverse effects are always unwanted, but unwanted effects are not necessarily adverse. An adverse effect from a prophylactic or therapeutic agent might be harmful or uncomfortable or risky. Side effects from chemotherapy include, but are not limited to, gastrointestinal toxicity such as, but not limited to, early and late-forming diarrhea and flatulence, nausea, vomiting, anorexia, leukopenia, anemia, neutropenia, asthenia, abdominal cramping, fever, pain, loss of body weight, dehydration, alopecia, dyspnea, insomnia, dizziness, mucositis, xerostomia, and kidney failure, as well as constipation, nerve and muscle effects, temporary or permanent damage to kidneys and bladder, flu-like symptoms, fluid retention, and temporary or permanent infertility. Side effects from radiation therapy include but are not limited to fatigue, dry mouth, and loss of appetite. Side effects from biological therapies/immunotherapies include but are not limited to rashes or swellings at the site of administration, flu-like symptoms such as fever, chills and fatigue, digestive tract problems and allergic reactions. Side effects from hormonal therapies include but are not limited to nausea, fertility problems, depression, loss of appetite, eye problems, headache, and weight fluctuation. Additional undesired effects typically experienced by patients are numerous and known in the art. Many are described in the Physicians' Desk Reference (56th ed., 2002).
[0061] By "therapeutically effective amount or dose" or "therapeutically sufficient amount or dose" or "effective or sufficient amount or dose" herein is meant a dose that produces therapeutic effects for which it is administered, in the context of the combination therapy in which it is administered. Often, the therapeutically effective or sufficient amount or dose of the compounds comprising the pharmaceutical compositions of the invention will be lower when administered in the specific combinations, than the doses that would be therapeutically effective or sufficient when the compounds are administered separately. The exact dose will depend on the purpose of the treatment, and will be ascertainable by one skilled in the art using known techniques (see, e.g., Lieberman, Pharmaceutical Dosage Forms (vols. 1-3, 1992); Lloyd, The Art, Science and Technology of Pharmaceutical Compounding (1999); Pickar, Dosage Calculations (1999); and Remington. The Science and Practice of Pharmacy, 20th Edition, 2003, Gennaro, Ed., Lippincott, Williams & Wilkins). In some embodiments, a therapeutically effective amount refers to that amount of the therapeutic agent sufficient to destroy, modify, control or remove primary, regional or metastatic cancer tissue. A therapeutically effective amount may refer to the amount of therapeutic agent sufficient to delay or minimize the spread of cancer. A therapeutically effective amount may also refer to the amount of the therapeutic agent that provides a therapeutic benefit in the treatment or management of cancer. Further, a therapeutically effective amount with respect to a therapeutic agent of the invention means that amount of therapeutic agent alone, or in combination with other therapies, that provides a therapeutic benefit in the treatment or management of cancer. In sensitized cells, the therapeutically effective dose can often be lower than the conventional therapeutically effective dose for non-sensitized cells. In some embodiments, a therapeutically effective amount refers to the amount of a therapeutic agent that, e.g., reduces the proliferation of cancer cells, increases the death of cancer cells or, reduces the size of a tumor or spread of a tumor in a subject. Preferably, a therapeutically effective amount of a therapeutic agent reduces the size of a tumor or the spread of a tumor in a subject 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%, ate 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 99% relative to a control such as PBS. In some embodiments, a therapeutically effective amount refers to the amount of a therapeutic agent that increases survival by 1 month, 2 months, 6 months, 1 year, 2 years, 3 years, 4 years, 5 years, 6 years, 7 years, 8 years, 9 years, 10 years, or more. In some embodiments, a therapeutically effective amount refers to the amount of a therapeutic agent that prevents the progression from PIN to prostate cancer.
[0062] The HDAC inhibitors encompassed by the methods and compositions of the present invention can be any known to those of skill in the art, including those described in Minucci et al, N ' ature 6:38-51 (2006). According to the invention, in some preferred embodiments the HDAC inhibitor is SFN, and in other embodiments the HDAC inhibitor is valproic acid. In yet other embodiments, the HDAC inhibitor may be TSA, SAHA, VPA derivatives, MS-275, clyclic hydroxamic acid-containing peptide, Apicidin, Trapoxin, or other HDAC inhibitors known by the skilled practitioner to be effective. Other HDAC inhibitors encompassed by the methods and compositions of the invention include the VPA derivatives as described in U.S. Patent Application Nos. 20050038113 to Groner, and 20040087652 to Gottlicher, as well as the compounds used to inhibit HDAC as disclosed in U.S. Patent Application Nos. 20070135438 to Payne, 20070060614 and 20070190022 to Bacopoulos, 20050107348 to Lan-Hargest, and 20070037738 to Hentsch, as well as U.S. Patent Nos. 7,169,801, 6,110,955, 6,905,669, and 7,126,001. Other HDAC inhibitors encompassed by the methods and compositions of the invention include the sulfonyl derivatives as described in U.S. Patent No. 7,205,304 to Van Emelen, the alpha-ketoepoxide compounds of U.S. Patent No. 7,057,057 to Lan- Hargest, the HDAC inhibitors based on trihalomethylcarbonyl compounds as described in U.S. Patent No. 7,193,105 to Lan-Hargest, and the HDAC inhibitors based on alpha- chalcogenmethylcarbonyl compounds of U.S. Patent No. 7,214,831 to Lan-Hargest. Each of the above patents and patent application publications is hereby incorporated by reference in its entirety.
[0063] In one aspect, the present invention provides a method of treating and methods of preventing prostate cancer, comprising administering to a subject a therapeutically effective amount of an HDAC inhibitor in combination with a therapeutically effective amount of an IGF-IR inhibitor, and/or an mTOR innhibitor. Any of the therapies may be further combined with androgen ablation therapy, such as bicalutamide. Various combinations of HDAC inhibitors and IGF-IR inhibitors, and/or mTOR inhibitors, as well as types of androgen ablation therapy, are contemplated as useful in treating prostate cancer.
[0064] In one embodiment, the daily dose of valproic acid is from about 15/mg/kg to about 60 mg/kg.
[0065] In one embodiment, the dose of valproic acid is sufficient to achieve from about 300 to about 1000 micromolar in patient serum. In another embodiment, the dose of valproic acid is sufficient to achieve from about 300 to about 867 micromolar in patient serum. In another embodiment, the dose of valproic acid is sufficient to achieve from about 500 to about 1000 micromolar in patient serum.
[0066] In some embodiments, the dose of SAHA is from about 200 mg/day to about 600 mg/day. In another embodiment, the dose of SAHA is about 400 mg/day.
[0067] In some embodiments, compounds are administered in combination, with ratios of those compounds which preserve the recommended daily doses of the compounds. In some embodiments, compounds are administered in combination, with ratios of those compounds which preserve the ranges of doses as described herein. [0068] The present invention also encompasses methods of treating and methods of preventing prostate cancer or PIN, methods of preventing the progression from PIN to prostate cancer, methods of preventing progression from low to higher grade prostate cancer, and methods of preventing recurrence of prostate cancer, comprising administering to a subject suffering therefrom a subject a therapeutically effective amount of an HDAC inhibitor in combination with a therapeutically effective amount of one or more of an IGF-IR inhibitor, and/or an mTOR inhibitor and optionally further in combination with androgen ablation therapy. Specifically, a preferred combination is VPA and EGCG for the treatment of pre-cancerous or early stage, androgen dependent prostate cancer.
[0069] The invention further contemplates methods of slowing the progression of naϊve (not yet treated with androgen ablation therapy) recurrent prostate cancer, comprising administering to a subject suffering therefrom a subject a therapeutically effective amount of an HDAC inhibitor in combination with a therapeutically effective amount of one or more of an IGF-IR inhibitor, and/or an mTOR inhibitor.
[0070] The invention further contemplates methods of preventing resistance to androgen ablation therapy, comprising administering to a subject suffering therefrom a subject a therapeutically effective amount of an HDAC inhibitor in combination with a therapeutically effective amount of one or more of an IGF-IR inhibitor, and/or an mTOR inhibitor, in addition to androgen ablation therapy.
[0071] In one embodiment, the combination comprises SFN, PPP, and rapamycin.
[0072] In one embodiment, the combination for treating a patient with AR-responsive prostate cancer comprises VPA and EGCG.
[0073] In one embodiment, the combination for treating a patient with AR independent (either AR positive or AR negative) prostate cancer comprises SFN and EGCG. In another embodiment, the combination for treating a patient with AR independent (either AR positive or AR negative) prostate cancer comprises SFN and PPP. In yet another embodiment, the combination for treating a patient with AR independent (either AR positive or AR negative) prostate cancer comprises SFN and rapamycin. In another embodiment, the combination for treating a patient with AR independent (either AR positive or AR negative) prostate cancer comprises EGCG, PPP and mTOR. [0074] The IGF-IR inhibitors contemplated in the methods and compositions of the present invention can be any known to one of skill in the art. The IGF-IR inhibitor may be for example, the green tea polyphenol, epigallocatechin gallate (EGCG), picropodophyllin (PPP), podophyllotoxin, podophyllotoxin derivatives, including those disclosed in U.S. Patent Application No. 20070123491 to Axelson, cyclolignans such as those disclosed in U.S. Patent Application No. 2004/0186169 to Larsson, and IGF-IR inhibitors such as those disclosed in U.S. Patent Application No. 20060193772 to Ochiai. Each of the above patent applications is hereby incorporated by reference in its entirety.
[0075] The mTOR inhibitors contemplated in the methods and compositions of the present invention can be any known to one of skill in the art. The mTOR inhibitor may be for example, temsirolimus, everolimus, rapamycin and rapamycin derivatives, including those rapamycin derivatives disclosed in U.S. Patent Application No. 20040147541 to Lane, which is hereby incorporated by reference in its entirety.
[0076] In one embodiment, particularly for late-stage, androgen-independent prostate cancer, the combination comprises SFN and rapamycin. In another embodiment, the combination comprises TSA and rapamycin. In yet another embodiment, the combination comprises VPA and rapamycin.
[0077] In one embodiment, the combination comprises SFN and EGCG. In another embodiment, the combination comprises TSA and EGCG. In yet another embodiment, the combination comprises VPA and EGCG.
[0078] In another embodiment, the combination comprises rapamycin and EGCG. In another embodiment, the combination comprises SFN, rapamycin, and EGCG. In another embodiment, the combination comprises TSA, rapamycin, and EGCG. In another embodiment, the combination comprises VPA, rapamycin, and EGCG.
[0079] In another embodiment, the combination comprises SFN and PPP. In another embodiment, the combination comprises TSA and PPP. In another embodiment, the combination comprises VPA and PPP. In another embodiment, the combination comprises rapamycin and PPP. In another embodiment, the combination comprises SFN, rapamycin, and PPP. In yet another embodiment, the combination comprises TSA, rapamycin, and PPP. In another embodiment, the combination comprises VPA, rapamycin, and PPP. [0080] In yet another embodiment, the combination comprises bicalutamide and VPA.
[0081] In one embodiment, the dose of rapamycin is from about 0.125 mg/day to about 1 mg/day.
[0082] In one preferred embodiment, the dose of valproic acid is between about 2g/day and about 3g/day.
[0083] In one embodiment, the daily dose of valproic acid is from about 15/mg/kg to about 60 mg/kg.
[0084] In one embodiment, the daily dose of valproic acid is sufficient to achieve about 300 to about 867 micromolar in patient serum. In another embodiment, the daily dose of valproic acid is sufficient to achieve about 300 to about 1000 micromolar in patient serum, and in another embodiment, the daily dose of valproic acid is sufficient to achieve about 500 to about 1000 micromolar in patient serum.
[0085] In some embodiments, the dose of SAHA is from about 200 mg/day to about 600 mg/day. In another embodiment, the dose of SAHA is about 400 mg/day.
[0086] In one embodiment, the dose of EGCG is from about 800 mg/day to about 1200 mg/day.
[0087] In another embodiment, the dose of EGCG is from about 300 mg/day to about 800 mg/day.
[0088] In one embodiment, the dose of rapamycin is from about 0.125 mg/day to about 1 mg/day.
[0089] In one embodiment, the combination comprises a ratio of VPA: EGCG from about 2 to about 3 parts VPA to about 1 part EGCG.
[0090] As relates to the inhibitors described herein, many different procedures can be used to specifically inactivate or silence a target gene or inhibit the activity of its gene product, as encompassed by the present invention. Inhibition of protein activity can be brought about at the level of gene transcription, protein translation or post-translational modifications. For instance, the activity of a protein can be inhibited by directly inhibiting the activity of the protein such as altering a catalytic domain or alternatively by reducing the amount of the protein in the cell by reducing the amount of mRNA encoding the protein. In each case, the level of protein activity in the cell is reduced. Various techniques can be used to knock down the activity of a protein and these include knockout technologies (antibodies, antisense RNA, and RNA interference) and compounds that specifically inhibit the protein activity.
[0091] In certain embodiments, an RNA interference (RNAi) molecule is used to decrease expression of a gene. RNA interference (RNAi) is defined as the ability of double-stranded RNA (dsRNA) to suppress the expression of a gene corresponding to its own sequence. RNAi is also called post-transcriptional gene silencing or PTGS. Since the only RNA molecules normally found in the cytoplasm of a cell are molecules of single- stranded mRNA, the cell has enzymes that recognize and cut dsRNA into fragments containing 21-25 base pairs (approximately two turns of a double helix). The antisense strand of the fragment separates enough from the sense strand so that it hybridizes with the complementary sense sequence on a molecule of endogenous cellular mRNA. This hybridization triggers cutting of the mRNA in the double-stranded region, thus destroying its ability to be translated into a polypeptide. Introducing dsRNA corresponding to a particular gene thus knocks out the cell's own expression of that gene in particular tissues and/or at a chosen time.
[0092] Double-stranded (ds) RNA can be used to interfere with gene expression in mammals (Wianny & Zemicka-Goetz, 2000, Nature Cell Biology 2: 70-75; incorporated herein by reference in its entirety). dsRNA is used as inhibitory RNA or RNAi of the function of the gene of interest to produce a phenotype that is the same as that of a null mutant of the gene of interest (Wianny & Zernicka-Goetz, 2000, Nature Cell Biology 2: 70-75).
[0093] Any therapy (e.g., chemotherapies, radiation therapies, hormonal therapies, and/or biological therapies/immunotherapies) which is known to be useful, or which has been used or is currently being used for the prevention, treatment, management or amelioration of cancer or one or more symptoms thereof can be used in accordance with the invention, and may be combined with any of the compositions described herein, and may encompass the other active ingredient described for some of the combination therapies herein.
[0094] In some embodiments, the anti-cancer agents contemplated in the methods and compositions of the present invention, which can be administered in combination with the compositions of the present invention include, but are not limited to suramin, estramustine, vinblastine, estramustine, etoposide, estramustine, Taxol, interferon, liarozole, prednisone, mitoxantrone, Paclitaxel, Docetaxel, Adriamycin, Provenge, and hormone-deprivation therapy.
[0095] It is within the scope of the present invention to treat many different types of subjects or patients, though preferably, the subject is a mammal. Preferred mammals include primates such as humans and chimpanzees, domestic animals such, as horses, cows, pigs, etc. and pets such as dogs and cats. Most preferably, the invention encompasses treating humans, and in particular, human males. The pharmaceutical compositions described herein may be used for the treatment of cancer, particularly for prostate cancer. The pharmaceutical compositions and methods of the present invention can be used to treat an individual with any type and/or stage of prostate cancer. There are several types of prostate cancer and there are several stages of prostate cancer, all of which are contemplated as treated by the methods and compositions of the present invention.
[0096] The present invention also contemplates all the different stages of prostate cancer, including stages I (A) through stages IV (D). In stage I, the tumor is so small that it cannot be felt by digital rectal examination (DRE). In stage II, the tumor is larger, and can be felt by DRE, but has not spread beyond the prostate. In stage III, the cancer has spread to the local tissue near the prostate. In stage IV, the cancer has spread to the lymph nodes or has metastasized to other body areas.
[0097] There are different types of staging. Clinical staging estimates how much cancer there is based on the results of the physical exam, imaging tests (x-rays, CT scans, etc.) and sometimes biopsies of affected areas. For certain cancers the results of other tests, such as blood tests, are also used in staging. Pathologic staging can only be done on patients who have had surgery to remove or explore the extent of the cancer. It combines the results of clinical staging (physical exam, imaging tests, etc.) with the results from the surgery. In some cases, the pathologic stage may be different from the clinical stage (for example, if the surgery shows the cancer is more extensive than it was previously thought to be). Restaging is sometimes used to determine the extent of the disease if a cancer recurs (comes back) after treatment.
[0098] In one embodiment, the methods and compositions of the present invention are used to treat patients with stage I prostate cancer. [0099] In one embodiment, the methods and compositions of the present invention are used to treat patients with stage II prostate cancer.
[00100] In one embodiment, the methods and compositions of the present invention are used to treat patients with stage III prostate cancer.
[00101] In one embodiment, the methods and compositions of the present invention are used to treat patients with stage IV prostate cancer, i.e. patients with metastatic cancer.
[00102] In another embodiment, the patient having prostate cancer has already failed other treatment regimens such as chemotherapy.
[00103] In one embodiment, the methods and pharmaceutical compositions of the present invention may be used to prevent the development of a cancer, particularly in an individual at higher risk than average to develop such cancer than other individuals, or to treat a patient afflicted with prostate cancer.
[00104] There are a number of ways to assess an individual's risk for prostate cancer, and any means of risk assessment is contemplated by the present invention as determining which subjects are at risk for prostate cancer and can undergo treatment via the methods and compositions of the present invention. The invention contemplates treatment for individuals with a higher than average lifetime risk for prostate cancer.
[00105] The invention provides methods treating asymptomatic patients who have a likelihood of benefiting from therapeutic treatment of prostate cancer. The asymptomatic patients can comprise patients in any of the many high risk groups for prostate cancer. Risk factors considered in preventing prostate cancer in subjects include, but are not limited to those diagnosed with prostatic intraepithelial neoplasia (PIN), prostate-specific antigen (PSA) levels, family history of prostate cancer (relatives with prostate cancer), age, age at which relatives were diagnosed, circulating hormone levels, genetic markers for prostate cancer such as PCA3 and 8q24, prostate stem cell antigen (PCSA), polymorphisms within the androgen receptor gene, levels of plasma hormones and sex hormone-binding globulin (SHBG), diet, obesity, race, ethnicity, nationality, whether the individual has had a vasectomy, and presence of prostatitis. The present invention also provides methods and compositions for preventing the progression of prostate cancer to a later stage for those who already have prostate cancer or precancerous indicators, as well as preventing the recurrence of prostate cancer for those in remission from prostate cancer. [00106] The combinations of the invention may be administered either prior to or following surgical removal of primary tumors and/or treatment such as administration of radiotherapy, surgery, or conventional chemotherapeutic drugs.
[00107] The methods and compositions of the present invention may be used advantageously in combination with any other treatment regimen for prostate cancer. Treatments for prostate cancer are well known in the art and continue to be developed. Treatments include but are not limited to surgery, including cryosurgery and radical surgery (prostatectomy), radiation therapy, transperineal brachytherapy, and chemotherapy. Any combination of therapies may be used in conjunction with the present invention.
[00108] The methods and compositions comprising the combination therapies described herein may also be used to reduce the proliferation of cancer cells, increase the death of cancer cells or, reduces the size of a tumor or spread of a tumor in a subject. It is contemplated by the present invention that the combination therapies described herein may reduce the size of a tumor or the spread of a tumor in a subject 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 99% relative to a control such as PBS. In some embodiments, the combination therapies described herein may increase survival by 1 month, 2 months, 6 months, 1 year, 2 years, 3 years, 4 years, 5 years, 6 years, 7 years, 8 years, 9 years, 10 years, or more, it may render the subject disease-free, or it may prevent the progression from PIN pr other precancerous stages to prostate cancer.
[00109] Pharmaceutically acceptable carriers are determined in part by the particular composition being administered, as well as by the particular method used to administer the composition. Accordingly, there are a wide variety of suitable formulations of pharmaceutical compositions of the present invention (see, e.g., Remington 's Pharmaceutical Sciences, 20th ed., 2003).
[00110] The compounds of the invention may be formulated into pharmaceutical compositions as natural or salt forms. Pharmaceutically acceptable non-toxic salts include the base addition salts (formed with free carboxyl or other anionic groups) which may be derived from inorganic bases such as, for example, sodium, potassium, ammonium, calcium, or ferric hydroxides, and such organic bases as isopropylamine, trimethylamine, 2-ethylamino-ethanol, histidine, procaine, and the like. Such salts may also be formed as acid addition salts with any free cationic groups and will generally be formed with inorganic acids such as, for example, hydrochloric, sulfuric, or phosphoric acids, or organic acids such as acetic, p-toluenesulfonic, methanesulfonic acid, oxalic, tartaric, mandelic, and the like. Salts of the invention include amine salts formed by the protonation of an amino group with inorganic acids such as hydrochloric acid, hydrobromic acid, hydroiodic acid, sulfuric acid, phosphoric acid, and the like. Salts of the invention also include amine salts formed by the protonation of an amino group with suitable organic acids, such as p-toluenesulfonic acid, acetic acid, and the like. Additional excipients which are contemplated for use in the practice of the present invention are those available to those of ordinary skill in the art, for example, those found in the United States Pharmacopeia Vol. XXII and National Formulary Vol. XVII, U.S. Pharmacopeia Convention, Inc., Rockville, Md. (1989), the relevant contents of which is incorporated herein by reference.
[00111] In a preferred embodiment, a composition of the invention is a pharmaceutical composition. Such compositions comprise a prophylactically or therapeutically effective amount of one or more prophylactic or therapeutic agents, including one or more of an HDAC inhibitor in combination with a therapeutically effective amount of one or more of an IGF-IR inhibitor, and/or an mTOR inhibitor, and optionally, an anti-androgen agent, and a pharmaceutically acceptable carrier.
[00112] In a specific embodiment, the term "pharmaceutically acceptable" means approved by a regulatory agency of the Federal or a state government or listed in the U.S. Pharmacopeia or other generally recognized pharmacopeia for use in animals, and more particularly in humans. The term "carrier" refers to a diluent, adjuvant (e.g., Freund's adjuvant (complete and incomplete)), excipient, or vehicle with which the therapeutic is administered. Such pharmaceutical carriers can be sterile liquids, such as water and oils, including those of petroleum, animal, vegetable or synthetic origin, such as peanut oil, soybean oil, mineral oil, sesame oil and the like. Water is a preferred carrier when the pharmaceutical composition is administered intravenously. Saline solutions and aqueous dextrose and glycerol solutions can also be employed as liquid carriers, particularly for injectable solutions. Suitable pharmaceutical excipients include starch, glucose, lactose, sucrose, gelatin, malt, rice, flour, chalk, silica gel, sodium stearate, glycerol monostearate, talc, sodium chloride, dried skim milk, glycerol, propylene, glycol, water, ethanol and the like. The composition, if desired, can also contain minor amounts of wetting or emulsifying agents, or pH buffering agents. These compositions can take the form of solutions, suspensions, emulsion, tablets, pills, capsules, powders, sustained-release formulations and the like. Oral formulation can include standard carriers such as pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharine, cellulose, magnesium carbonate, etc. Examples of suitable pharmaceutical carriers are described in "Remington's Pharmaceutical Sciences" by E. W. Martin. Such compositions will contain a prophylactically or therapeutically effective amount of a prophylactic or therapeutic agent preferably in purified form, together with a suitable amount of carrier so as to provide the form for proper administration to the patient. The formulation should suit the mode of administration. In a preferred embodiment, the pharmaceutical compositions are sterile and in suitable form for administration to a subject, preferably an animal subject, more preferably a mammalian subject, and most preferably a human subject.
[00113] In a specific embodiment, it may be desirable to administer the pharmaceutical compositions of the invention locally to the area in need of treatment; this may be achieved by, for example, and not by way of limitation, local infusion, by injection, or by means of an implant, said implant being of a porous, non-porous, or gelatinous material, including membranes, such as sialastic membranes, or fibers. Preferably, when administering one or more prophylactic or therapeutic agents, care must be taken to use materials to which the prophylactic or therapeutic agents do not absorb.
[00114] In another embodiment, the composition can be delivered in a vesicle, in particular a liposome (see Langer, Science 249:1527-1533 (1990); Treat et al., in Liposomes in the Therapy of Infectious Disease and Cancer, Lopez-Berestein and Fidler (eds.), Liss, New York, pp. 353-365 (1989); Lopez-Berestein, pp. 317-327; see generally above.).
[00115] In yet another embodiment, the composition can be delivered in a controlled release or sustained release system. In one embodiment, a pump may be used to achieve controlled or sustained release (see Langer, supra; Sefton, 1987, CRC Crit. Ref. Biomed. Eng. 14:20; Buchwald et al., 1980, Surgery 88:507; Saudek et al., 1989, N. Engl. J. Med. 321 :574). In another embodiment, polymeric materials can be used to achieve controlled or sustained release of the antibodies of the invention or fragments thereof (see e.g., Medical Applications of Controlled Release, Langer and Wise (eds.), CRC Pres., Boca Raton, FIa. (1974); Controlled Drug Bioavailability, Drug Product Design and Performance, Smolen and Ball (eds.), Wiley, N.Y. (1984); Ranger and Peppas, 1983, J., Macromol. Sci. Rev. Macromol. Chem. 23:61; see also Levy et al., 1985, Science 228:190; During et al., 1989, Ann. Neural. 25:351; Howard et al., 1989, J. Neurosurg. 7 1 :105); U.S. Pat. No. 5,679,377; U.S. Pat. No. 5,916,597; U.S. Pat. No. 5,912,015; U.S. Pat. No. 5,989,463; U.S. Pat. No. 5,128,326; International Publication No. WO 99/15154; and International Publication No. WO 99/20253. Examples of polymers used in sustained release formulations include, but are not limited to, poly(2-hydroxy ethyl methacrylate), poly(methyl methacrylate), poly(acrylic acid), poly(ethylene-co-vinyl acetate), poly(methacrylic acid), polyglycolides (PLG), polyanhydrides, poly(N-vinyl pyrrolidone), poly(vinyl alcohol), polyacrylamide, poly(ethylene glycol), polylactides (PLA), poly(lactide-co-glycolides) (PLGA), and polyorthoesters. In a preferred embodiment, the polymer used in a sustained release formulation is inert, free of leachable impurities, stable on storage, sterile, and biodegradable. In yet another embodiment, a controlled or sustained release system can be placed in proximity of the therapeutic target, i.e., the lungs, thus requiring only a fraction of the systemic dose (see, e.g., Goodson, in Medical Applications of Controlled Release, supra, vol. 2, pp. 115-138 (1984)).
[00116] Controlled release systems are discussed in the review by Langer (1990, Science 249:1527-1533). Any technique known to one of skill in the art can be used to produce sustained release formulations comprising one or more antibodies of the invention or fragments thereof. See, e.g., U.S. Pat. No. 4,526,938, International publication No. WO 91/05548, International publication No. WO 96/20698, Ning et al., 1996, "Intratumoral Radioimmunotheraphy of a Human Colon Cancer Xenograft Using a Sustained-Release Gel," Radiotherapy & Oncology 39:179-189, Song et al., 1995, "Antibody Mediated Lung Targeting of Long-Circulating Emulsions," PDA Journal of Pharmaceutical Science & Technology 50:372-397, Cleek et al., 1997, "Biodegradable Polymeric Carriers for a bFGF Antibody for Cardiovascular Application," Pro. Int'l. Symp. Control. ReI. Bioact. Mater. 24:853-854, and Lam et al., 1997, "Microencapsulation of Recombinant Humanized Monoclonal Antibody for Local Delivery," Proc. Int'l. Symp. Control ReI. Bioact. Mater. 24:759-760, each of which is incorporated herein by reference in their entirety. [00117] In a specific embodiment where the composition of the invention is one or more nucleic acid molecules encoding one or more prophylactic or therapeutic agents, the nucleic acid can be administered in vivo to promote expression of its encoded prophylactic or therapeutic agents, by constructing it as part of an appropriate nucleic acid expression vector and administering it so that it becomes intracellular, e.g., by use of a retroviral vector (see U.S. Pat. No. 4,980,286), or by direct injection, or by use of microparticle bombardment (e.g., a gene gun; Biolistic, Dupont), or coating with lipids or cell-surface receptors or transfecting agents, or by administering it in linkage to a homeobox-like peptide which is known to enter the nucleus (see e.g., Joliot et al., 1991, Proc. Natl. Acad. Sci. USA 88:1864-1868), etc. Alternatively, a nucleic acid can be introduced intracellularly and incorporated within host cell DNA for expression by homologous recombination.
[00118] A pharmaceutical composition of the invention is formulated to be compatible with its intended route of administration. Examples of suitable routes of administration include, but are not limited to, parenteral (e.g., intravenous, intramuscular, intradermal, intra-tumoral, intra-synovial, and subcutaneous), oral (e.g., inhalation), intranasal, transdermal (topical), transmucosal, intra-tumoral, intra-synovial, vaginal, and rectal administration. In a specific embodiment, the composition is formulated in accordance with routine procedures as a pharmaceutical composition adapted for intravenous, subcutaneous, intramuscular, oral, intra-tummoral, intra synnovial, intranasal or topical administration to human beings. Typically, compositions for intravenous administration are solutions in sterile isotonic aqueous buffer. Where necessary, the composition may also include a solubilizing agent and a local anesthetic such as lignocaine to ease pain at the site of the injection.
[00119] If the compositions of the invention are to be administered topically, the compositions can be formulated in the form of, e.g., a toothpaste, ointment, cream, transdermal patch, lotion, gel, oral gel, shampoo, spray, aerosol, solution, emulsion, or other form well-known to one of skill in the art. See, e.g., Remington's Pharmaceutical Sciences and Introduction to Pharmaceutical Dosage Forms, 4.sup.th ed., Lea & Febiger, Philadelphia, Pa. (1985). For non-sprayable topical dosage forms, viscous to semi-solid or solid forms comprising a carrier or one or more excipients compatible with topical application and having a dynamic viscosity preferably greater than water are typically employed. Suitable formulations include, without limitation, solutions, suspensions, emulsions, creams, ointments, powders, liniments, salves, and the like, which are, if desired, sterilized or mixed with auxiliary agents (e.g., preservatives, stabilizers, wetting agents, buffers, or salts) for influencing various properties, such as, for example, osmotic pressure. Other suitable topical dosage forms include sprayable aerosol preparations wherein the active ingredient, preferably in combination with a solid or liquid inert carrier, is packaged in a mixture with a pressurized volatile (e.g., a gaseous propellant, such as freon), or in a squeeze bottle. Moisturizers or humectants can also be added to pharmaceutical compositions and dosage forms if desired. Examples of such additional ingredients are well-known in the art.
[00120] If the compositions of the invention are to be administered intranasally, the compositions can be formulated in an aerosol form, spray, mist or in the form of drops. In particular, prophylactic or therapeutic agents for use according to the present invention can be conveniently delivered in the form of an aerosol spray presentation from pressurized packs or a nebuliser, with the use of a suitable propellant, e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas. In the case of a pressurized aerosol the dosage unit may be determined by providing a valve to deliver a metered amount. Capsules and cartridges of, e.g., gelatin for use in an inhaler or insufflator may be formulated containing a powder mix of the compound and a suitable powder base such as lactose or starch.
[00121] If the compositions of the invention are to be administered orally, the compositions can be formulated orally in the form of, e.g., gum, tablets, capsules, cachets, gelcaps, solutions, suspensions and the like. Tablets or capsules can be prepared by conventional means with pharmaceutically acceptable excipients such as binding agents (e.g., pregelatinised maize starch, polyvinylpyrrolidone or hydroxypropyl methylcellulose); fillers (e.g., lactose, microcrystalline cellulose or calcium hydrogen phosphate); lubricants (e.g., magnesium stearate, talc or silica); disintegrants (e.g., potato starch or sodium starch glycolate); or wetting agents (e.g., sodium lauryl sulphate). The tablets may be coated by methods well-known in the art. Liquid preparations for oral administration may take the form of, for example, solutions, syrups or suspensions, or they may be presented as a dry product for constitution with water or other suitable vehicle before use. Such liquid preparations may be prepared by conventional means with pharmaceutically acceptable additives such as suspending agents (e.g., sorbitol syrup, cellulose derivatives or hydrogenated edible fats); emulsifying agents (e.g., lecithin or acacia); non-aqueous vehicles (e.g., almond oil, oily esters, ethyl alcohol or fractionated vegetable oils); and preservatives (e.g., methyl or propyl-p-hydroxybenzoates or sorbic acid). The preparations may also contain buffer salts, flavoring, coloring and sweetening agents as appropriate. Preparations for oral administration may be suitably formulated for slow release, controlled release or sustained release of a prophylactic or therapeutic agent(s).
[00122] The compositions of the invention may be formulated for parenteral administration by injection, e.g., by bolus injection or continuous infusion. Formulations for injection may be presented in unit dosage form, e.g., in ampoules or in multi-dose containers, with an added preservative. The compositions may take such forms as suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents. Alternatively, the active ingredient may be in powder form for constitution with a suitable vehicle, e.g., sterile pyrogen-free water, before use.
[00123] The compositions of the invention may also be formulated in rectal compositions such as suppositories or retention enemas, e.g., containing conventional suppository bases such as cocoa butter or other glycerides.
[00124] In addition to the formulations described previously, the compositions of the invention may also be formulated as a depot preparation. Such long acting formulations may be administered by implantation (for example subcutaneously or intramuscularly) or by intramuscular injection. Thus, for example, the compositions may be formulated with suitable polymeric or hydrophobic materials (for example as an emulsion in an acceptable oil) or ion exchange resins, or as sparingly soluble derivatives, for example, as a sparingly soluble salt.
[00125] The compositions of the invention can be formulated as neutral or salt forms. Pharmaceutically acceptable salts include those formed with anions such as those derived from hydrochloric, phosphoric, acetic, oxalic, tartaric acids, etc., and those formed with cations such as those derived from sodium, potassium, ammonium, calcium, ferric hydroxides, isopropylamine, triethylamine, 2-ethylamino ethanol, histidine, procaine, etc.
[00126] Generally, the ingredients of compositions of the invention are supplied either separately or mixed together in unit dosage form, for example, as a dry lyophilized powder or water free concentrate in a hermetically sealed container such as an ampoule or sachette indicating the quantity of active agent. Where the composition is to be administered by infusion, it can be dispensed with an infusion bottle containing sterile pharmaceutical grade water or saline. Where the composition is administered by injection, an ampoule of sterile water for injection or saline can be provided so that the ingredients may be mixed prior to administration.
[00127] Kits
[00128] The invention provides a pharmaceutical pack or kit comprising one or more containers filled with individual components (in pharmaceutical formulations) of the combination therapies described herein; for example, contained filled with one or more prophylactic or therapeutic agents, including one or more of an HDAC inhibitor in combination with a therapeutically effective amount of one or more of an IGF-IR inhibitor, and/or an mTOR inhibitor, and optionally, an anti-androgen agent, as well as other active ingredients, such as chemotherapy agents. The pharmaceutical pack or kit may further comprises one or more other prophylactic or therapeutic agents useful for the treatment of a disease or disorder. The invention also provides a pharmaceutical pack or kit comprising one or more containers filled with one or more of the ingredients of the pharmaceutical compositions of the invention. Optionally associated with such container(s) can be a notice in the form prescribed by a governmental agency regulating the manufacture, use or sale of pharmaceuticals or biological products, which notice reflects approval by the agency of manufacture, use or sale for human administration.
[00129] Articles of Manufacture
[00130] The present invention also encompasses a finished packaged and labeled pharmaceutical product. This article of manufacture includes the appropriate unit dosage form in an appropriate vessel or container such as a glass vial or other container that is hermetically sealed. In the case of dosage forms suitable for parenteral administration the active ingredient is sterile and suitable for administration as a particulate free solution. In other words, the invention encompasses both parenteral solutions and lyophilized powders, each being sterile, and the latter being suitable for reconstitution prior to injection. Alternatively, the unit dosage form may be a solid suitable for oral, transdermal, intratumoral, intra-synovial, topical or mucosal delivery. [00131] In a specific embodiment, the unit dosage form is suitable for intravenous, intramuscular, intratumoral, intra-synovial, or subcutaneous delivery. Thus, the invention encompasses solutions, preferably sterile, suitable for each delivery route.
[00132] As with any pharmaceutical product, the packaging material and container are designed to protect the stability of the product during storage and shipment. Further, the products of the invention include instructions for use or other informational material that advise the physician, technician or patient on how to appropriately prevent or treat the disease or disorder in question. In other words, the article of manufacture includes instruction means indicating or suggesting a dosing regimen including, but not limited to, actual doses, monitoring procedures (such as methods for monitoring mean absolute lymphocyte counts, tumor cell counts, calcium concentration, and tumor size) and other monitoring information.
[00133] More specifically, the invention provides an article of manufacture comprising packaging material, such as a box, bottle, tube, vial, container, sprayer, insufflator, intravenous (i.v.) bag, envelope and the like; and at least one unit dosage form of a pharmaceutical agent contained within said packaging material.
[00134] In a specific embodiment, an article of manufacture comprises packaging material and a pharmaceutical agent and instructions contained within said packaging material, wherein said pharmaceutical agent comprises at least one or more prophylactic or therapeutic agents, including one or more of an HDAC inhibitor in combination with a therapeutically effective amount of one or more of an IGF-IR inhibitor, and/or an mTOR inhibitor, and optionally, an anti-androgen agent and a pharmaceutically acceptable carrier, and said instructions indicate a dosing regimen for preventing, treating or managing a subject with cancer.
[00135] In therapeutic use for the treatment of cancer, the compounds utilized in the pharmaceutical method of the invention are administered at the initial dosage of about 0.001 mg/kg to about 1000 mg/kg daily. A daily dose range of about 0.01 mg/kg to about 500 mg/kg, or about 0.1 mg/kg to about 200 mg/kg, or about 1 mg/kg to about 100 mg/kg, or about 10 mg/kg to about 50 mg/kg, can be used. The dosages, however, may be varied depending upon the requirements of the patient, the severity of the condition being treated, and the compound being employed. For example, dosages can be empirically determined considering the type and stage of cancer diagnosed in a particular patient. The dose administered to a patient, in the context of the present invention should be sufficient to effect a beneficial therapeutic response in the patient over time. The size of the dose also will be determined by the existence, nature, and extent of any adverse side-effects that accompany the administration of a particular compound in a particular patient. Determination of the proper dosage for a particular situation is within the skill of the practitioner. Generally, treatment is initiated with smaller dosages which are less than the optimum dose of the compound. Thereafter, the dosage is increased by small increments until the optimum effect under circumstances is reached. For convenience, the total daily dosage may be divided and administered in portions during the day, if desired. Doses can be given daily, or on alternate days, as determined by the treating physician.
[00136] Characterization and Demonstration of Therapeutic or Prophylactic Utility
[00137] Toxicity and efficacy of the prophylactic and/or therapeutic treatments and protocols of the instant invention can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., for determining the LD50 (the dose lethal to 50% of the population) and the ED50 (the dose therapeutically effective in 50% of the population). The dose ratio between toxic and therapeutic effects is the therapeutic index and it can be expressed as the ratio LD50/ED50. Prophylactic and/or therapeutic agents that exhibit large therapeutic indices are preferred. While prophylactic and/or therapeutic agents that exhibit toxic side effects may be used, care should be taken to design a delivery system that targets such agents to the site of affected tissue in order to minimize potential damage to uninfected cells and, thereby, reduce side effects.
[00138] The data obtained from the cell culture assays and animal studies can be used in formulating a range of dosage of the prophylactic and/or therapeutic agents for use in humans. The dosage of such agents lies preferably within a range of circulating concentrations that include the ED50 with little or no toxicity. The dosage may vary within this range depending upon the dosage form employed and the route of administration utilized. For any agent used in the method of the invention, the therapeutically effective dose can be estimated initially from cell culture assays. A dose may be formulated in animal models to achieve a circulating plasma concentration range that includes the IC50 (i.e., the concentration of the test compound that achieves a half- maximal inhibition of symptoms) as determined in cell culture. Such information can be used to more accurately determine useful doses in humans. Levels in plasma may be measured, for example, by high performance liquid chromatography. [00139] In order to determine therapeutic or prophylactic utility, it is encompassed by the present invention to use any of the assays described herein, including those described and illustrated in the Examples section below, as well as those known in the art. Also encompassed by the invention to determine therapeutic or prophylactic utility are any relevant cancer, and more specifically, prostate cancer animal models. For example, one may utilize the TRAMP model that utilizes the minimal rat probasin promoter to express the SV40 early genes (T and t antigens; Tag) as well as a number of transgenic lines using the long probasin promoter to express large T antigen, collectively termed the 'LADY' model. The Cryptdin-2-T and Gg-SV40 T models also develop progressive prostate cancer and may be used in the invention. Further encompassed by the invention, pending safety and efficacy, are clinical trials to assess the combinations of the present invention.
EXAMPLES
[00140] It is understood that the following examples and embodiments described herein are for illustrative purposes only and that various modifications or changes in light thereof will be suggestive to persons skilled in the art and are to be included within the spirit and purview of this application and the scope of the appended claims. All publications, patents, and patent applications cited herein are hereby incorporated by reference in their entirety for all purposes.
[00141] The Examples demonstrate the following results, illustrated in FIGS 1-4:
[00142] The HDAC inhibitor TSA combines effectively to reduce proliferation of IGF- IR inhibitors (EGCG and PPP) and the mTOR inhibitor rapamycin in prostate cancer cells. In androgen-insensitive DU 145 cells the combination of TSA, PPP and rapamycin is more efficacious than any one of the drugs alone or any two of these drugs used in combination. TSA also combines effectively with PPP in androgen-sensitive LNCaP cells.
[00143] The HDAC inhibitor VPA combines effectively to reduce proliferation of IGF-IR inhibitors (EGCG and PPP) and the mTOR inhibitor rapamycin in prostate cancer cells in a cell type specific manner. In androgen-sensitive LNCaP cells, the combination of VPA and EGCG is more efficacious than either drug used alone. VPA also combines effectively with rapamycin to inhibit LNCaP cell proliferation better than either drug used alone. [00144] In addition, in androgen-insensitive DU 145 cells, SFN and rapamycin combine effectively to inhibit cell proliferation, and a triple combination of SFN with an IGF-IR inhibitor (PPP or EGCG) and rapamycin is more efficacious than any one of these drugs alone or any two drugs in combination.
[00145] VPA combines effectively with bicalutamide to reduce proliferation in androgen-sensitive prostate cells. A dose of 5OnM bicalutamide was chosen as an effective dose to inhibit androstendiol-stimulated cell proliferation. VPA inhibited cell proliferation by itself and also increased the efficacy of bicalutamide.
[00146] The anti-androgen, bicalutamide combines effectively with VPA and EGCG to inhibit androgen-sensitive prostate cell proliferation. A dose of 1 μM bicalutamide was chosen as an effective dose to inhibit androstenediol-stimulated cell proliferation (see FIG. 4A). The triple combination of VPA, EGCG and bicalutamide is more effective at inhibiting androgen-sensitive prostate cell proliferation than any one of these drugs or double combination of the drugs. Bicalutamide and VPA combine effectively to inhibit androgen-sensitive prostate cell proliferation, as do bicalutamide and EGCG (see FIG. 4B).
EXAMPLE 1 Effect of cocktail treatments on cellular proliferation in LNCaP cells
[00147] The effect of the HDAC inhibitors, SFN, TSA, and VPA alone or in combination with rapamycin, further in combination with either the IGF-IR inhibitor, EGCG or PPP on cell proliferation of the human prostate cancer cell line, LNCaP after being in culture for 6 days. Proliferation was measured using a DNA-binding fluorescent dye. Cells were treated with the following doses of inhibitors: 500μM VPA, 2nM TSA, 1 μM SFN, 7μM EGCG, 3OnM PPP, 2nM rapamycin (see FIG. 1).
EXAMPLE 2 Effect of cocktail treatments on cellular proliferation in DU 145 cells
[00148] The effect of the HDAC inhibitors, SFN, TSA, and VPA alone or in combination with rapamycin, further in combination with either of the IGF-IR inhibitors EGCG or PPP on cell proliferation of the human prostate cancer cell line, DU145, after being in culture for 6 days. Proliferation was measured using a DNA-binding fluorescent dye. Cells were treated with the following doses of inhibitors: 500μM VPA, 2nM TSA, 1 μM SFN, 7μM EGCG, 3OnM PPP, 2nM rapamycin (see FIG. 2).
EXAMPLE 3 Effect of cocktail treatments on cellular proliferation in PC3 cells
[00149] The effect of the HDAC inhibitors, SFN, TSA, and VPA alone or in combination with rapamycin, further in combination with either if the IGF-IR inhibitors EGCG or PPP on cell proliferation of the human prostate cancer cell line, PC3, after being in culture for 6 days. Proliferation was measured using a DNA-binding fluorescent dye. Cells were treated with the following doses of inhibitors: 500μM VPA, 2nM TSA, 1 μM SFN, 7μM EGCG, 3OnM PPP, 2nM rapamycin (see FIG. 3).
EXAMPLE 4 Effect of cocktail treatments with bicalutamide on cellular proliferation in LNCaP cells
[00150] The effects of the anti-androgen, bicalutamide, the HDAC inhibitor, VPA, and the IGF-IR inhibitor EGCG, alone and in double or triple combination on cell proliferation of the human prostate cancer cell line, LNCaP after being in culture for 7 days. LNCaP cells were cultured in 5% Charcoal Dextran Stripped Serum (CDSS) RPMI+50nM Androstenediol. Proliferation was measured using a DNA-binding fluorescent dye. A dose of 5OnM bicalutamide was chosen as an effective dose to inhibit androstendiol-stimulated cell proliferation (see FIG. 4).
[00151] It is understood that the examples and embodiments described herein are for illustrative purposes only and that various modifications or changes in light thereof will be suggested to persons skilled in the art and are to be included within the spirit and purview of this application and scope of the appended claims. All publications, patents, and patent applications cited herein are hereby incorporated by reference in their entirety for all purposes.

Claims

What is claimed is:
1. A method of preventing progression from PIN to prostate cancer, comprising administering to a subject in need thereof a therapeutically effective amount of an HDAC inhibitor in combination an IGF-IR inhibitor.
2. The method of claim 1, wherein the HDAC inhibitor is VPA and the IGF-IR inhibitor is EGCG.
3. The method of claim 1 , wherein the HDAC inhibitor is VPA.
4. The method of claim 1, wherein the IGF-IR is EGCG.
5. A method of preventing progression from low to higher grade prostate cancer, comprising administering to a subject in need thereof a therapeutically effective amount of an HDAC inhibitor in combination an IGF-IR inhibitor.
6. The method of claim 5, wherein the HDAC inhibitor is VPA and the IGF-IR inhibitor is EGCG.
7. The method of claim 5, wherein the HDAC inhibitor is VPA.
8. The method of claim 5, wherein the IGF-IR is EGCG.
9. A method of preventing recurrence of prostate cancer, comprising administering to a subject in need thereof a therapeutically effective amount of an HDAC inhibitor in combination with an IGF-IR inhibitor.
10. The method of claim 9, wherein the HDAC inhibitor is VPA and the IGF-IR inhibitor is EGCG.
11. A method of slowing progression of naϊve, recurrent prostate cancer that has not yet treated with androgen ablation, comprising administering to a subject in need thereof a therapeutically effective amount of one or more of an HDAC inhibitor, an IGF-IR inhibitor, an mTOR inhibitor, or combinations thereof.
12. The method of claim 11 , wherein the HDAC inhibitor is VPA.
13. The method of claim 11, wherein the HDAC inhibitor is SFN.
14. The method of claim 11, wherein the IGF-IR inhibitor is EGCG.
15. The method of claim 11, wherein the IGF-IR inhibitor is PPP.
16. The method of claim 11 , wherein the mTOR inhibitor is rapamycin.
17. A method of preventing resistance to androgen ablation therapy in a subject with prostate cancer, comprising administering to said subject a therapeutically effective amount of one or more of an HDAC inhibitor, an IGF-IR inhibitor, an mTOR inhibitor, or combinations thereof.
18. The method of claim 17, wherein the HDAC inhibitor is selected from the group consisting of VPA, SFN, and TSA.
19. The method of claim 17, wherein the IGF-IR inhibitor is EGCG.
20. The method of claim 17, wherein the IGF-IR inhibitor is PPP.
21. The method of claim 17, wherein the mTOR inhibitor is rapamycin.
22. A method of treating androgen independent prostate cancer comprising administering to a subject in need thereof a therapeutically effective amount of one or more of an HDAC inhibitor, an IGF-IR inhibitor, an mTOR inhibitor, or combinations thereof.
23. The method of claim 22, comprising administering SFN and rapamycin.
24. A method of treating or preventing androgen-dependent prostate cancer comprising administering to a subject in need thereof, a combination of VPA and EGCG.
25. A method of treating or preventing androgen-dependent prostate cancer comprising administering to a subject in need thereof, a combination of an anti-androgen therapy and VPA.
26. The method of claim 25, wherein the anti-androgen therapy is bicalutamide.
27. A method of treating or preventing androgen-independent prostate cancer comprising administering to a subject in need thereof, a combination of SFN and EGCG.
28. A method of treating or preventing androgen-independent prostate cancer comprising administering to a subject in need thereof, a combination of SFN and PPP.
29. A method of treating or preventing prostate cancer comprising administering to a subject in need thereof, a combination of rapamycin, PPP, and SFN.
30. A method of preventing progression from PIN to prostate cancer, comprising administering to a subject in need thereof a therapeutically effective amount of an VPA in combination with EGCG.
31. A method of preventing progression from low to higher grade prostate cancer, comprising administering to a subject in need thereof a therapeutically effective amount of VPA in combination with EGCG.
32. A method of preventing recurrence of prostate cancer, comprising administering to a subject in need thereof a therapeutically effective amount of VPA in combination with EGCG.
33. A method of treating androgen independent prostate cancer comprising administering to a subject in need thereof a therapeutically effective amount of SFN in combination with rapamycin.
34. A method of treating or preventing androgen-dependent prostate cancer comprising administering to a subject in need thereof, a combination of one or more of an anti- androgen therapy, an HDAC inhibitor, and an IGF-IR inhibitor.
35. The method of claim 34, wherein the anti-androgen therapy is bicalutamide.
36. The method of claim 34, wherein the HDAC inhibitor is VPA.
37. The method of claim 34, wherein the IGF-IR inhibitor is EGCG.
38. The method of claim 34, wherein the anti-androgen therapy is bicalutamide and the HDAC inhibitor is VPA.
39. The method of claim 34, wherein the anti-androgen therapy is bicalutamide and the HDAC inhibitor is SFN.
40. The method of claim 34, wherein the anti-androgen therapy is bicalutamide and the IGF-IR inhibitor is EGCG.
41. The method of claim 34, wherein the anti-androgen therapy is bicalutamide, the HDAC inhibitor is VPA, and the IGF-IR inhibitor is EGCG.
42. The method of claim 34, wherein the anti-androgen therapy is bicalutamide, the HDAC inhibitor is SFN, and the IGF-IR inhibitor is EGCG.
PCT/US2009/035464 2008-02-27 2009-02-27 Combination therapy for prostate cancer WO2009108857A2 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US3190308P 2008-02-27 2008-02-27
US61/031,903 2008-02-27
US3393108P 2008-03-05 2008-03-05
US61/033,931 2008-03-05

Publications (2)

Publication Number Publication Date
WO2009108857A2 true WO2009108857A2 (en) 2009-09-03
WO2009108857A3 WO2009108857A3 (en) 2010-01-14

Family

ID=41016726

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2009/035464 WO2009108857A2 (en) 2008-02-27 2009-02-27 Combination therapy for prostate cancer

Country Status (1)

Country Link
WO (1) WO2009108857A2 (en)

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2010001096A2 (en) * 2008-07-01 2010-01-07 Provexis Natural Products Limited Treatment
EP2285406A2 (en) * 2008-05-05 2011-02-23 The Regents of the University of California Inhibitory effects of nordihydroguaiaretic acid (ndga) on the igf-1 receptor and androgen dependent growth of lapc-4 prostate cancer cells
CN104117064A (en) * 2013-04-28 2014-10-29 无锡杰西医药科技有限公司 Combined application of isothiocyanate compound and hormone medicine
US9381246B2 (en) 2013-09-09 2016-07-05 Triact Therapeutics, Inc. Cancer therapy
EP3034076A4 (en) * 2013-08-13 2016-10-19 Jc Wuxi Company Inc Combined application of isothiocyanate compound and anti-cancer medicine
WO2017034234A1 (en) * 2015-08-21 2017-03-02 서울대학교 산학협력단 Composite formulation for treating cancer having hdac inhibitor resistance
US9834575B2 (en) 2013-02-26 2017-12-05 Triact Therapeutics, Inc. Cancer therapy
CN108159419A (en) * 2017-12-27 2018-06-15 中国药科大学 Induce application of the substance of IRF8 expression in the drug for preparing treatment androgen receptor unconventionality expression associated diseases
WO2021127305A1 (en) * 2019-12-20 2021-06-24 Virginia Commonwealth University Methods and compositions for inducing apoptosis in cancer stem cells

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040186169A1 (en) * 2001-06-19 2004-09-23 Olle Larsson Use of specific cyclolignans
WO2005111232A2 (en) * 2004-05-17 2005-11-24 MAX-PLANCK-Gesellschaft zur Förderung der Wissenschaften e.V. Silencing of tumor-suppressive genes by cpg-methylation in prostate cancer
WO2006017904A1 (en) * 2004-08-19 2006-02-23 Purdue Research Foundation Improved anticancer treatment
US20070123580A1 (en) * 2003-05-21 2007-05-31 Atadja Peter W Combination of histone deacetylase inhibitors with chemotherapeutic agents
US20070292512A1 (en) * 2006-06-09 2007-12-20 Merrion Research Ii Limited Solid Oral Dosage Form Containing an Enhancer

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040186169A1 (en) * 2001-06-19 2004-09-23 Olle Larsson Use of specific cyclolignans
US20070123580A1 (en) * 2003-05-21 2007-05-31 Atadja Peter W Combination of histone deacetylase inhibitors with chemotherapeutic agents
WO2005111232A2 (en) * 2004-05-17 2005-11-24 MAX-PLANCK-Gesellschaft zur Förderung der Wissenschaften e.V. Silencing of tumor-suppressive genes by cpg-methylation in prostate cancer
WO2006017904A1 (en) * 2004-08-19 2006-02-23 Purdue Research Foundation Improved anticancer treatment
US20070292512A1 (en) * 2006-06-09 2007-12-20 Merrion Research Ii Limited Solid Oral Dosage Form Containing an Enhancer

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
ATTARD ET AL.: 'Improving the outcomes of patients with castration-resistant prostate cancer through rational drug development.' BRITISH J CANCER vol. 95, 2006, pages 767 - 774 *

Cited By (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2285406A2 (en) * 2008-05-05 2011-02-23 The Regents of the University of California Inhibitory effects of nordihydroguaiaretic acid (ndga) on the igf-1 receptor and androgen dependent growth of lapc-4 prostate cancer cells
JP2011519925A (en) * 2008-05-05 2011-07-14 ザ リージェンツ オブ ザ ユニヴァーシティー オブ カリフォルニア Inhibitory effect of nordihydroguaiaretic acid (NDGA) on IGF-1 receptor and androgen-dependent proliferation of LAPC-4 prostate cancer cells
EP2285406A4 (en) * 2008-05-05 2012-05-02 Univ California Inhibitory effects of nordihydroguaiaretic acid (ndga) on the igf-1 receptor and androgen dependent growth of lapc-4 prostate cancer cells
WO2010001096A2 (en) * 2008-07-01 2010-01-07 Provexis Natural Products Limited Treatment
WO2010001096A3 (en) * 2008-07-01 2010-04-08 Provexis Natural Products Limited Treatment
US9834575B2 (en) 2013-02-26 2017-12-05 Triact Therapeutics, Inc. Cancer therapy
CN104117064A (en) * 2013-04-28 2014-10-29 无锡杰西医药科技有限公司 Combined application of isothiocyanate compound and hormone medicine
EP3034076A4 (en) * 2013-08-13 2016-10-19 Jc Wuxi Company Inc Combined application of isothiocyanate compound and anti-cancer medicine
US9381246B2 (en) 2013-09-09 2016-07-05 Triact Therapeutics, Inc. Cancer therapy
WO2017034234A1 (en) * 2015-08-21 2017-03-02 서울대학교 산학협력단 Composite formulation for treating cancer having hdac inhibitor resistance
CN108159419A (en) * 2017-12-27 2018-06-15 中国药科大学 Induce application of the substance of IRF8 expression in the drug for preparing treatment androgen receptor unconventionality expression associated diseases
WO2021127305A1 (en) * 2019-12-20 2021-06-24 Virginia Commonwealth University Methods and compositions for inducing apoptosis in cancer stem cells

Also Published As

Publication number Publication date
WO2009108857A3 (en) 2010-01-14

Similar Documents

Publication Publication Date Title
WO2009108857A2 (en) Combination therapy for prostate cancer
US11215617B2 (en) Treatment of metastatic prostate cancer
US20080085874A1 (en) Small molecule potentiator of hormonal therapy for breast cancer
TWI402064B (en) Cancer therapy
KR20180096621A (en) Combination for the treatment of cancer
WO2007041282A2 (en) Methods and compositions for treatment of cystic fibrosis
US20190262319A1 (en) Novel methods for treating cancer
Xu et al. 2-Methoxy-5 ((3, 4, 5-trimethosyphenyl) seleninyl) phenol inhibits MDM2 and induces apoptosis in breast cancer cells through a p53-independent pathway
Tao et al. Role of S100A3 in human hepatocellular carcinoma and the anticancer effect of sodium cantharidinate
CN111494605B (en) Medical application of CREG protein in preventing or treating doxorubicin myocardial injury
ES2905360T3 (en) Compositions and methods for detecting, treating and preventing diseases and disorders
WO2016179002A1 (en) Compositions and methods for treatment of cancer
US20220218658A1 (en) Use of delta-tocotrienol in th prevention of cancer and cancer recurrence
WO2009108361A1 (en) Methods for treatment and prevention of breast cancer
WO2019051025A2 (en) Treatment for aggressive cancers by targeting c9orf72
WO2009142968A2 (en) Valproic acid salts
EP3850100A1 (en) Targeting egln1 in cancer
EP3656783A1 (en) Peptide derivative and pharmaceutical composition containing same
US20130157959A1 (en) Use of hades as tumor suppressor target
US20240009174A1 (en) Methods for disrupting mitochondrial unfolded protein response
US20240068037A1 (en) Methods and compositions for diagnosing and treating prostate cancer based on long noncoding rna overlapping the lck gene that regulates prostate cancer cell growth
US20230321035A1 (en) Lasofoxifene treatment of aromatase-resistant er+ cancer
Xie et al. Goserelin combined with bicalutamide improves immune function and inflammatory response in patients with advanced prostate cancer
JP2022514654A (en) Treatment for squamous cell carcinoma
US20220054610A1 (en) Slow-cycling cell-rna based nanoparticle vaccine to treat cancer

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: 09714382

Country of ref document: EP

Kind code of ref document: A2

NENP Non-entry into the national phase in:

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 09714382

Country of ref document: EP

Kind code of ref document: A2