US20190380998A1 - Methods of treating and preventing melanoma with s-equol - Google Patents

Methods of treating and preventing melanoma with s-equol Download PDF

Info

Publication number
US20190380998A1
US20190380998A1 US16/384,428 US201916384428A US2019380998A1 US 20190380998 A1 US20190380998 A1 US 20190380998A1 US 201916384428 A US201916384428 A US 201916384428A US 2019380998 A1 US2019380998 A1 US 2019380998A1
Authority
US
United States
Prior art keywords
equol
cells
tumor
antibody
formulation
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US16/384,428
Inventor
Rong Li
Bin Yuan
Tyler Curiel
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
University of Texas System
Original Assignee
University of Texas System
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 University of Texas System filed Critical University of Texas System
Priority to US16/384,428 priority Critical patent/US20190380998A1/en
Publication of US20190380998A1 publication Critical patent/US20190380998A1/en
Assigned to THE BOARD OF REGENTS OF THE UNIVERSITY OF TEXAS SYSTEM reassignment THE BOARD OF REGENTS OF THE UNIVERSITY OF TEXAS SYSTEM ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: CURIEL, TYLER, LI, RONG, YUAN, BIN
Priority to US17/819,863 priority patent/US20220387380A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/35Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having six-membered rings with one oxygen as the only ring hetero atom
    • A61K31/352Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having six-membered rings with one oxygen as the only ring hetero atom condensed with carbocyclic rings, e.g. methantheline 
    • A61K31/3533,4-Dihydrobenzopyrans, e.g. chroman, catechin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/2818Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against CD28 or CD152
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/2827Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against B7 molecules, e.g. CD80, CD86

Definitions

  • This invention relates to methods and compositions for treating or preventing melanoma with a pharmaceutically effective amount of S-equol or a pharmaceutical composition comprising S-equol.
  • the method relates to a combination therapy using S-equol and another anti-cancer treatment, such as immunotherapy.
  • the methods and compositions of the present invention are suitable for various types of cancers, particularly those responsive to targeting of the estrogen receptor beta (ER ⁇ ).
  • ER ⁇ estrogen receptor beta
  • the methods and compositions of the present invention are suitable for treating melanoma.
  • Malignant melanoma is a potentially serious skin cancer that results from the uncontrolled growth of pigment cells, called melanocytes.
  • Melanoma may be the result of UV-induced changes to the skin, and its occurrence may be increased as a result of various genetic mutations. Mutations arising in the c-Kit gene, the BRAF gene, the CDKN2A gene, which regulates cell division, the MDm2 gene, which is a negative regulator of the p53 tumor suppressor gene, the retinoblastoma RB1 gene, and the MC1R (melanocortin-1-receptor) gene are among those that may increase the risk of developing melanoma. Treatment of melanoma may include the use of surgery, chemotherapy and/or immunotherapy. (Gupta et al., 2016; Clark et al., 2016; Steinberg et al., 2017).
  • PD-1 is a checkpoint protein on T-cells that serves as an “off switch” to keep T cells from attacking other cells in the body.
  • PD-1 interacts with PD-L1 on tumor cells.
  • PD-L1 interacts with PD-1, it prevents the T-cells from attacking the tumor cells.
  • PD-1 inhibitors are currently being used to treat melanoma and other cancers.
  • CD4 + T cells play a key role in the functioning of a healthy immune system. They assist B cells to make antibodies, activate the microbe killing capacity of macrophages and recruit other immune cells to infected or inflamed areas of the body. These activities are orchestrated through their production of various cytokines and chemokines. It has been known for some time that uncommitted CD4 + T-cells can differentiate into Th1 or Th2 cells, based on the prevailing pro-inflammatory/anti-inflammatory environment, and that these activated Th1 and Th2 cells had distinct cytokine production patterns and functions. Generally, Th1 cells were associated with the eradication of intracellular pathogens whereas Th2 cells were heavily involved in responses against extracellular pathogens and parasites.
  • Th17 cells which have a pro-inflammatory bias was identified.
  • Subsequent research using animal models and human studies has demonstrated a key role for Th17 cells in the immune system's defense against extracellular bacteria and fungi as well as the development of autoimmune diseases, mediated by the secretion of IL-17 by these cells.
  • CD8 + (cytotoxic) T cells like CD4 + Helper T cells, are generated in the thymus and express the T cell receptor. However, rather than the CD4 molecule, cytotoxic T cells express a dimeric coreceptor, CD8, usually composed of one CD8a and one CD8P chain. CD8 + T cells recognize peptides presented by MHC Class I molecules, found on all nucleated cells. The CD8 heterodimer binds to a conserved portion (the ⁇ 3 region) of MHC Class I during T cell/antigen presenting cell interactions (see FIG. 1 ). CD8 + T cells (often called cytotoxic T lymphocytes, or CTLs) are very important for immune defense against intracellular pathogens, including viruses and bacteria, and for tumor surveillance.
  • CTLs cytotoxic T lymphocytes
  • cytotoxic granules When a CD8 + T cell recognizes its antigen and becomes activated, it has three major mechanisms to kill infected or malignant cells. The first is secretion of cytokines, primarily TNF- ⁇ and IFN- ⁇ , which have anti-tumor and anti-microbial effects. The second major function is the production and release of cytotoxic granules. These granules, also found in natural killer (NK) cells, contain two families of proteins, perforin, and granzymes. Perforin forms a pore in the membrane of the target cell, similar to the membrane attack complex of complement. This pore allows the granzymes also contained in the cytotoxic granules to enter the infected or malignant cell.
  • NK natural killer
  • Granzymes are serine proteases which cleave the proteins inside the cell, shutting down the production of viral proteins and ultimately resulting in apoptosis of the target cell.
  • the cytotoxic granules are released only in the direction of the target cell, aligned along the immune synapse, to avoid non-specific bystander damage to healthy surrounding tissue (see FIG. 1 ).
  • CD8 + T cells are able to release their granules, kill an infected cell, then move to a new target and kill again, often referred to as serial killing.
  • the third major function of CD8 + T cell destruction of infected cells is via Fas/FasL interactions.
  • CD8 + T cells express FasL on the cell surface, which binds to its receptor, Fas, on the surface of the target cell. This binding causes the Fas molecules on the surface of the target cell to trimerize, which pulls together signaling molecules. These signaling molecules result in the activation of the caspase cascade, which also results in apoptosis of the target cell. Because CD8 + T cells can express both molecules, Fas/FasL interactions are a mechanism by which CD8 + T cells can kill each other, called fratricide, to eliminate immune effector cells during the contraction phase at the end of an immune response.
  • CD8 + T cells can also contribute to an excessive immune response that leads to immunopathology, or immune-mediated damage.
  • Bitesized Immunology https://www.immunology.org/public-information/bitesized-immunology/cells/cd8-t-cells.
  • the PK136 monoclonal antibody reacts with mouse NK1.1, an antigen expressed by natural killer cells and a subset of T cells in the NK1.1 mouse strains including C57BL and NZB.
  • mouse NK1.1 an antigen expressed by natural killer cells
  • a subset of T cells in the NK1.1 mouse strains including C57BL and NZB.
  • Several commonly used laboratory mouse strains such as BALB/c, SJL, AKR, CBA, C3H and A do not express the NK1.1 antigen.
  • DXS 14-5971 is used for detection of NK cells in these strains.
  • Simultaneous staining of C57BL/6 spleen cells with PK136 and DXS reveals coexpression of both markers by a majority of cells as well as presence of small populations of DXS+PK136 ⁇ and DXS ⁇ PK136+ cells.
  • CD45 lymphocyte common antigen
  • CD45 is a receptor-linked protein tyrosine phosphatase that is expressed on all leucocytes, and which plays a crucial role in the function of these cells.
  • the extracellular domain of CD45 is expressed in several different isoforms, and the particular isoform(s) expressed depends on the particular subpopulation of cell, their state of maturation, and whether or not they have previously been exposed to antigen. It has been established that the expression of CD45 is essential for the activation of T cells via the TCR, and that different CD45 isoforms display a different ability to support T cell activation.
  • CD45 tyrosine phosphatase activity of the intracellular region of CD45 has been shown to be crucial for supporting signal transduction from the TCR, the nature of the ligands for the different isoforms of CD45 have been elusive. Moreover, the precise mechanism by which potential ligands may regulate CD45 function is unclear. Interestingly, in T cells CD45 has been shown to associate with numerous molecules, both membrane associated and intracellular; these include components of the TCR-CD3 complex and CD4/CD8. In addition, CD45 is reported to associate with several intracellular protein tyrosine kinases including R561ck and p59fyn of the src family, and ZAP-70 of the Syk family, and with numerous proteins of 29-34 kDa.
  • CD45-associated molecules may play an important role in regulating CD45 tyrosine phosphatase activity and function.
  • CD45-associated molecules e.g. CD45-AP and LPAP
  • PD-1 and its ligand PD-L1 are immune checkpoint molecules that dampen T cell immunity (Lin et al. (2010); Curiel et al. (2003); Brahmer et al. (2012); Pardoll and Drake (2012); Pardoll (2012)).
  • PD-L1 is overexpressed in tumor cells and some immune cells, including Tregs (Lin et al. (2010); Curiel et al. (2003); Pardoll (2012); Topalian et al. 2012)).
  • Immunotherapy with anti-PD-1 ( ⁇ PD-1) and anti-PD-LI antibodies ( ⁇ PD-L1) has proven successful for treating various cancers (Brahmer (2012); Pardoll and Drake (2012); Pardoll (2012)).
  • ER ⁇ receptors There are two distinct types of estrogen receptors that mediate diverse physiological effects of estrogens on breast cancer cells.
  • the first receptor is the ER ⁇ receptor which supports estrogen-dependent breast tumor growth.
  • the second is the ER ⁇ receptor which appears to work in an opposite fashion to significantly attenuate the growth of breast tumor cells in preclinical models (Clarke et al. (2003); Deroo and Korach (2006); Deroo and Buensuceso (2010); Honma et al (2008); Katzenellenbogen and Katzenellenbogen (2000); McDonnell and Norris (2002); Murphy and Watson (2006); Shaaban et al (2008).
  • ER ⁇ can be viewed in as a tumor suppressor gene in breast cancer.
  • ER ⁇ is expressed in about 40% of TNBC cancers, and the therapeutic potentials of targeting ER ⁇ have not yet been investigated in TNBC.
  • Two recent advances have now made it feasible to move from the preclinical models and into clinical research.
  • the first advance is a newly discovered mechanism of rallying ER ⁇ 's antitumor activity (Harris (2007); Hartman et al (2009); Heldring et al. (2007); Thomas and Gustafsson (2011)), and the second is the development of an oral formulation of an ER ⁇ agonist (S-equol) that has already been tested for clinical safety, pharmacodynamics and tolerance in humans (Jackson et al. (2011a & b); Setchell et al. (20005); Setchell (2002); Schwen et al. (2012).
  • Estrogen receptor (ER ⁇ ; nuclear or cytoplasmic) is reported to be present in approximately half of TNBC (Marotti et al. (2010); Reese et al. (2014)) as well as in melanoma (Marzagalli et al., 2016).
  • ER ⁇ in host cells has also been implicated in melanoma tumor inhibition (Cho et al. (2010)).
  • Cho et al. showed that syngeneic murine melanoma cells grafted to recipient ER ⁇ -KO animals grew more robustly than those in WT recipient.
  • TMA Prognostic Tissue Microarray
  • NCI National Cancer Institute
  • pY36-negative tumors were found to have statistically significant shorter disease-free and overall survival than those with pY36-positive tumors.
  • the association with survival was only seen in Stage II & III disease, which raises the intriguing possibility that pY36 activity may have an effect on disease progression from locally advanced to metastatic breast cancer.
  • pY36 intensity appears to have a stronger correlation with patient outcome than total ER ⁇ , underscoring the clinical importance of this previously unappreciated phosphotyrosine switch. See, FIG. 3 .
  • S-equol can be produced either chemically (i.e., chemical synthesis) or by biotransformation (biosynthesis) through the metabolism of daidzein, an isoflavone found in soy and red clover, by gut bacteria.
  • biotransformation biosynthesis
  • the structure of S-equol is shown below.
  • Equol has a chiral center and therefore can exist in two enantiomeric forms.
  • S-equol, R-equol, racemic equol, and non-racemic mixtures of equol (collectively “equol”); compositions of equol; anhydrous crystalline polymorph of equol; processes for the preparation of equol; and methods of using equol are described in U.S. Pat. No. 8,716,497 (filed Sep. 10, 2012); U.S. Pat. No. 8,048,913 (filed Sep. 14, 2009); U.S. Pat. No. 7,960,432 (filed Jul. 3, 2008); U.S. Pat. No. 7,396,855 (filed Jul.
  • Formulations comprising isoflavones and products derived therefrom have been used in the past to treat disease.
  • a mixture of equol, genistein, and daidzein, or a mixture of equol, genistein, daidzein, and IBSO03569 have shown potential for treating or preventing neurodegeneration and Alzheimer's disease. See Zhao et al. (2009); U.S. Pat. No. 8,552,057; Yao et al. (2013) (collectively “Brinton et al.”). S-equol alone has also been described for treating Alzheimer's Disease. See, U.S. application Ser. No. 15/659,114 published as U.S. Patent Publication 2018/0028491, and International Patent Publication WO/2018/022604, which are hereby incorporated by reference in their entireties.
  • S-equol preferably pure and isolated S-equol
  • S-equol in combination with immunotherapy is particularly effective in treating melanoma.
  • the invention also relates to compositions comprising S-equol as described for the methods herein.
  • FIG. 1 Schematic depiction of the action of CD8 + cells.
  • FIG. 2 Schematic depiction of the action of CD4 + cells.
  • FIG. 3 Association of pY36 with disease outcomes in breast cancer.
  • FIG. 3A shows the Kaplan-Meier estimate of disease-free and overall survival in Stages II-III specimens correlating with IHC intensity of pY36-ER ⁇ .
  • FIG. 3B shows the Kaplan-Meier estimate of disease-free and overall survival in Stages II-III specimens correlating with IHC intensity of (A) and total ER ⁇ IHC.
  • FIG. 4 A schema for combination therapy. Mice were administered one of: (1) vehicle+rat IgG2a; (2) S-equol+rat IgG2a; (3) vehicle+anti-PD-1 antibody; or (4) S-equol+anti-PD-1 antibody. Mice receiving anti-PD-1 antibody were given 250 ⁇ g every 3 days, intraperitoneally, and mice receiving S-equol were given 50 mg/kg per day by oral gavage. Treatment started 7 days after tumor challenge with B16 melanoma.
  • FIG. 5 A role of ER ⁇ signaling in immune cells.
  • FIG. 5A shows the schema for the chimera experiments.
  • FIG. 5C shows total CD8 + T cells from tumors.
  • FIG. 5D shows interferon gamma (IFN- ⁇ ) expressing CD8 + T cells from tumors.
  • IFN- ⁇ interferon gamma
  • FIG. 6 Effect of S-equol in combination with an anti-PD-1 antibody on mice challenged with an B16 melanoma tumor cell line over 12 days. Mice were administered anti-PD-1 antibody at 250 ⁇ g every 3 days, intraperitoneally, and S-equol at 50 mg/kg per day by oral gavage.
  • FIG. 6 shows that separately, S-equol and anti-PD-1 antibody reduce tumor volume compared to control, but S-equol in combination with an anti-PD-1 antibody reduces tumor volume compared to S-equol or anti-PD-1 antibody alone over 12 days post-challenge.
  • FIG. 7 Effect of S-equol in combination with an anti-PD-1 antibody on mice challenged with an B16 melanoma tumor cell line over 19 days. Mice were administered anti-PD-1 antibody at 250 ⁇ g every 3 days, intraperitoneally, and S-equol at 50 mg/kg per day by oral gavage.
  • FIG. 7 shows that separately, S-equol and anti-PD-1 antibody reduce tumor volume compared to control, but S-equol in combination with an anti-PD-1 antibody reduces tumor volume compared to S-equol or anti-PD-1 antibody alone over 19 days post-challenge.
  • FIG. 8 Effect of S-equol in combination with an anti-PD-1 antibody on tumor weight in mice challenged with an B16 melanoma tumor cell line.
  • FIG. 8A shows that separately, S-equol and anti-PD-1 antibody reduce tumor weight compared to control, but S-equol in combination with an anti-PD-1 antibody reduces tumor weight compared to S-equol or anti-PD-1 antibody alone.
  • FIG. 8B shows mouse total body weight for each treatment.
  • FIG. 9 Effect of S-equol in combination with an anti-PD-1 antibody and LY500307 on mice challenged with an B16 melanoma tumor cell line over 16 days. Mice were administered anti-PD-1 antibody at 250 ⁇ g every 3 days, intraperitoneally, and S-equol at 50 mg/kg per day by oral gavage.
  • FIG. 9A shows that separately, S-equol and anti-PD-1 antibody reduce tumor volume compared to control, but S-equol in combination with an anti-PD-1 antibody reduces tumor volume compared to S-equol or anti-PD-1 antibody alone over 12 days post-challenge.
  • FIG. 9B shows results with LY500307, a selective ER ⁇ agonist (SERBA-1; Erteberel®).
  • FIG. 10 Generalized protocol for human clinical trial with S-equol.
  • substantially pure means about 90% pure, preferably 95% pure, and more preferably 98% pure of any contaminating proteins.
  • substantially free means about less than about 10%, preferably less than 5%, and more preferably less than 2% of any contaminating proteins.
  • Approximately or “about” means within +/ ⁇ 10%, preferably within +/ ⁇ 5%, more preferably within +/ ⁇ 2% of that which is being measured.
  • the present invention relates to the prevention and/or treatment of melanoma with S-equol.
  • Dosage amounts and administration schedules for S-equol will depend on whether the S-equol is being administered prophylactically to a patient at risk of developing melanoma, or being administered as treatment for a patient already diagnosed with melanoma.
  • a person at risk for developing melanoma may be a person with a family history of melanoma, and/or may have one or more mutations in a CDKN2 A gene, MDm2 gene, RB1 gene, or MC1R gene.
  • dosages and administration schedules may also vary depending on the stage of the cancer (stage I, stage II, stage III, stage IV or stage V), the number of lymph nodes involved, tumor size and the availability of and/or decision to co-administer other therapies. Dosages and administration schedules may also vary depending on various molecular markers present on or in tumor cells, including, but not limited to Hsp90, RGS1, osteopontin, HER3, ING4, ING3, NCOA3, MCM4, MCM6, Ki67 and pY36. Diagnostic tests for markers which are prognostic for the aggressiveness and/or likelihood of recurrence of a tumor may be used as a factor or determining dosage regimens. (Gogas et al. (2009), incorporated by reference in its entirety for all purposes.)
  • S-equol can be administered one or more times per day at 1-400 mg per dose, more preferably 10-320 mg, more preferably 50-150 mg.
  • Non-limiting examples include 2 mg, 5 mg, 10 mg, 15 mg, 20 mg, 40 mg, 50 mg, 80 mg, 100 mg, 150 mg, 160 mg, 200 mg, 250 mg, 300 mg 320 mg, etc. or approximately or about those doses.
  • the dose may be administered one, two, three or four times per day, preferably twice per day (B.I.D.)
  • the regimen can be continued indefinitely, or for such time in intervals where markers are examined and responsiveness to the dose is determined. Examples of such intervals are two weeks, four weeks, two months, four months, six months, etc. or about or approximately those intervals. No upper limit, with respect to administration schedule, is required.
  • the S-equol administered is preferably formulated for oral administration; however, other routes of administration are also contemplated, including rectal, optical, buccal (for example sublingual), parenteral (for example subcutaneous, intramuscular, intradermal and intravenous) and transdermal administration.
  • routes of administration including rectal, optical, buccal (for example sublingual), parenteral (for example subcutaneous, intramuscular, intradermal and intravenous) and transdermal administration.
  • compositions or formulations according to the present invention can comprise one or more pharmaceutically-acceptable or industrial standard fillers.
  • the filler must not be deleterious to a subject treated with the composition.
  • the filler can be solid or a liquid, or both.
  • the filler can be formulated with the active S-equol as a unit-dose, for example a tablet, which can typically contain from about 10% to 80% by weight of S-equol.
  • Compositions can be prepared by any of the well known techniques of pharmacy, for example admixing the components, optionally including excipients, diluents (for example water) and auxiliaries as are well known in the pharmaceutical field.
  • compositions suitable for oral administration can be presented in discrete units, such as capsules, cachets, lozenges, or tablets, each containing a predetermined amount of the extract; as a powder or granules; as a solution or a suspension in an aqueous or non-aqueous liquid; or as an oil-in-water or water-in-oil emulsion.
  • Such compositions can be prepared by any suitable method of pharmacy which includes the step of bringing into association the active S-equol and one or more suitable carriers (which can contain one or more accessory ingredients as noted above).
  • compositions of the invention are prepared by uniformly and intimately admixing the S-equol with a liquid or finely divided solid carrier, or both, and then, if necessary, shaping the resulting mixture.
  • a tablet can be prepared by comprising or moulding a powder or granules containing the extract, optionally with one or more accessory ingredients.
  • Compressed tablets can be prepared by compressing in a suitable machine, the extracts in the form of a powder or granules optionally mixed with a binder, lubricant, inert diluents, and/or surface active/dispersing agent(s).
  • Moulded tablets can be made by moulding, in a suitable machine, the powdered compound moistened with an inert liquid binder.
  • Suitable fillers such as sugars, for example lactose, saccharose, mannitol or sorbitol, cellulose preparations and/or calcium phosphates, for example tricalcium phosphate or calcium hydrogen phosphate, and also binders such as starch pastes using, for example, corn, wheat, rice or potato starch, gelatin, tragacanth, methylceullose and/or polyvinylpyrrolidone, and, if desired, disintegrators, such as the above-mentioned starches, also carboxymethyl starch, cross linked polyvinyl pyrrolidone, agar or alginic acid or a salt thereof, such as sodium alginate.
  • sugars for example lactose, saccharose, mannitol or sorbitol
  • cellulose preparations and/or calcium phosphates for example tricalcium phosphate or calcium hydrogen phosphate
  • binders such as starch pastes using, for example, corn, wheat, rice or potato
  • Excipients can be flow conditioners and lubricants, for example silicic acid, talc, stearic acid or salts thereof, such as magnesium or calcium stearate, and/or polyethylene glycol.
  • Dragee cores are provided with suitable, optionally enteric, coatings, there being used, inter alia, concentrated sugar solutions which can comprise gum arabic, talc, polyvinylpyrrolidone, polyethylene glycol and/or titanium dioxide, or coating solutions in suitable organic solvents or solvent mixtures, or, for the preparation of enteric coatings, solutions of suitable cellulose preparations, such as microcrystalline cellulose, acetylcellulose phthalate or hydroxypropylmethylcellulose phthalate. Dyes or pigments can be added to the tablets or dragee coatings, for example for identification purposes or to indicate different doses of active ingredients.
  • compositions are dry-filled capsules made, for example, of gelatin, and soft, sealed capsules made of gelatin and a plasticiser, such as glycerol or sorbitol.
  • the dry-filled capsules can comprise the extracts in the form of granules, for example in admixture with fillers, such as lactose, binders, such as starches, and/or glicants, such as talc or magnesium stearate, and, where appropriate, stabilizers.
  • the extract is preferably dissolved or suspended in suitable liquids, such as fatty oils, paraffin oil or liquid polyethylene glycols, to which stabilizers can also be added.
  • compositions comprising S-equol include those described in U.S. Pat. No. 7,960,432 (filed Jul. 3, 2008); U.S. Pat. No. 7,396,855 (filed Jul. 24, 2003); and U.S. Pat. No. 9,408,824 (filed Mar. 5, 2014)—the disclosures of each are hereby incorporated by reference in their entireties.
  • S-equol can be prepared chemically (i.e., chemical synthesis) according to the processes described in U.S. Pat. No. 8,716,497 (filed Sep. 10, 2012); U.S. Pat. No. 8,263,790 (filed Jun. 1, 2011); U.S. Pat. No. 7,960,573 (filed May 4, 2009); U.S. Pat. No. 7,528,267 (filed Aug. 1, 2005) and U.S. Pat. No. 9,914,718 (filed Oct. 14, 2014)—the disclosures of each are hereby incorporated by reference in their entireties.
  • S-equol can be enantioselectively prepared using an iridium catalyst with a chiral ligand. These methods of enantioselectively preparing S-equol are incorporated by reference.
  • S-equol can be a single anhydrous crystalline polymorph of S-equol, such as the anhydrous crystalline polymorph of S-equol described in U.S. Pat. No. 9,914,718 (application Ser. No. 14/883,617, filed Oct. 14, 2015)—the disclosure of which, including the chemical and physical properties used to characterize the anhydrous crystalline polymorph of S-equol, is incorporated by reference in their entireties.
  • 2016/0102070 has the following characteristic X-ray powder diffraction pattern wavenumbers (cm ⁇ 1 ): 3433, 3023, 3003, 2908, 2844, 1889, 1614, 1594, 1517, 1508, 1469, 1454, 1438, 1400, 1361, 1323, 1295, 1276, 1261, 1234, 1213, 1176, 1156, 1116, 1064, 1020, 935, 897, 865, 840, 825, 810, 769, 734, 631, 616, 547, 517, 480, and 461.
  • the characterizations of anhydrous crystalline polymorph of S-equol are incorporated by reference.
  • the S-equol can be administered in combination with one or more additional cancer treatments, including surgery, cytotoxic chemotherapy, radiation, immunotherapy, cancer vaccines, inhibitors of cellular pathways (protein or peptide, nucleic acid-based [antisense oligonucleotides including DNA oligonucleotides, antisense siRNA, shRNA] and/or hormonal (adjuvant endocrine) therapy.
  • cytotoxic chemotherapy suitable for treating melanoma include, but are not limited to imiquimod cream (Zyclara®), dacarbazine (DTIC) and temozolomide (Temodar®).
  • immunotherapy examples include but are not limited to nivolumab (Opdivo®), pembrolizumab (Keytruda®), nivolumab (Opdivo®), and ipilirurab (Yervoy®), imatinib (Glevac®), nilotinib (Tasigna®) or interferon and interleukin-2 (IL-2).
  • T-VEC vaccine Imlygic®
  • BCG Bacille Calmette-Guerin
  • Appropriate dosages of immunotherapy monoclonal antibodies or other cancer therapeutic are determined based on the indication, and their determination is well within the skill in the art. Dosing may be in mg, in mg/kg or mg/m 2 . See, Sachs et al. (2016), which is incorporated by reference herein in its entirety for all purposes, including examples of therapeutic antibodies, small molecules and dosages.
  • the S-equol and/or additional therapeutic composition may be administered once, twice, three times or more per day, depending on the dose, toxicity and indication. Dosing may occur over several days, weeks or months, and may be continuous or intermittent. Typical dosages for immunotherapeutics are 50-500 mg/day, more preferably 100-400 mg/day, more preferably 150-250 mg/day.
  • the dose may be 150 mg/day or 150 mg twice per day, or 250 mg/day or 250 mg twice a day.
  • the dosage may be given based on patient weight, for instance 0.5-10 mg/kg, more preferably about 1, 2 or 5 mg/kg.
  • Dosage may also be given as mg/m 2 , for example 50-500 mg/m 2 , more preferably 100-300 mg/m 2 , for example about 100, 150, 200, 250 or 300 mg/m 2 .
  • the dosages may be about or greater than the lower end of any of the afore-stated ranges, or about or less than the upper end of any of the afore-stated ranges.
  • Responsiveness to the S-equol+/ ⁇ other cancer therapy can be assessed by analyzing one or more cellular markers, by imaging techniques, by cellular proliferation assays and by direct examination of tumors.
  • the analysis of cellular markers can be done with immunohistochemical or immunocytochemical techniques or by nucleic acid detection, such as hybridization techniques and polymerase chain reaction (PCR).
  • PCR polymerase chain reaction
  • a whole-body knock-in (K1) mouse model (C57BL/6) was established in which the corresponding tyrosine residue of endogenous mouse ER ⁇ is mutated to phenylalanine (Y-F). Consistent with previously reported ER ⁇ knockout (KO) mice (Krege et al. (1998)), these phosphorylation mutant KI mice had no overt developmental defects and were grossly indistinguishable from their WT littermates (unpublished data).
  • a mouse chimera experiment was conducted involving a bone marrow transplant.
  • WT recipient mice were first irradiated (10 Gy) to kill endogenous bone marrow cells, followed by transplant with bone marrow from syngeneic KI or WT donors. After confirming successful chimerism in KI>WT and WT>WT mice, tumor cells were injected 8 weeks after bone marrow transplant.
  • tumor-infiltrating immune cell populations were analyzed.
  • Total numbers of tumor-infiltrating CD4 + and CD8 + T cells were reduced in KI>WT versus WT>WT mice ( FIG. 5C and data not shown), further supporting the notion that antitumor immunity in KI mice is compromised.
  • the prevalence of IFN ⁇ -producing CD8 + (antitumor) cells was significantly lower in tumors from KI>WT versus WT>WT mice ( FIG. 5D ), suggesting compromised cytotoxic potency of CD8+ T cells in the absence of functional ER ⁇ signaling.
  • Melanoma cell line B16 was tested to determine the effects of a PD-1 inhibitor, S-equol and a combination of the two therapies on tumor growth.
  • This experimental design includes four arms: (1) controls, (2) ⁇ PD-1, (3) S-equol, and (4) ⁇ PD-1+S-equol ( FIG. 4 ) by injecting into mice 5 ⁇ 10 5 cells subcutaneously.
  • Antibodies were administered ⁇ -PD-1: 250 ⁇ g/mice by i.p. injection every 3 days and S-equol was administered at 50 mg/kg/day by oral gavage. Treatment began 7 day after tumor challenge. Tumor growth trajectories were compared within treated mice using a repeated measures linear mixed model.
  • tumors In addition to measurements of tumor size and weight in each arm, immunophenotyping is performed to gain more mechanistic insight into the antitumor effects of mono- and combinational therapies.
  • Tumors, spleens, and draining lymph nodes are harvested and weighed. Anti-coagulated blood was collected by cardiac puncture. Tumors are paraffin-embedded for immunohistochemistry and snap-frozen for mRNA and protein analysis by Luminex. Flow cytometry is used (for phenotype/functions) and ViCell (for quantification) to analyze immune cells from tumor infiltrates, tumor-draining lymph nodes, and spleens.
  • Analyses include CD3 + total T cells and CD4 + and CD8 + T cell subsets; effector function (e.g., IFN- ⁇ , TNF- ⁇ , IL-2, perforin, CD107a); activation (e.g., CD69, CD44, CD62L); and exhaustion (e.g., PD-1, Tim3, Lag3).
  • effector function e.g., IFN- ⁇ , TNF- ⁇ , IL-2, perforin, CD107a
  • activation e.g., CD69, CD44, CD62L
  • exhaustion e.g., PD-1, Tim3, Lag3
  • Antigen-presenting cells CD11b + CD1c ⁇ monocyte/macrophages, CD11b + CD11c+ dendritic cells), NK1.1 + NKp46 + NK cells, regulatory T cells (Treg, CD3 + CD4 + CD25 hi Foxp3 + ), and myeloid-derived suppressor cells (MDSC, CD11b + Gr ⁇ 1 hi ) are also analyzed.
  • CD8 + T cell receptor diversity were assessed with a commercial kit (Adaptive Biotechnologies). Local cell proliferation is also tested with Ki67 and BrdU stain.
  • Tumors harvested from mice are fixed in 10% neutral-buffered formalin, dehydrated, embedded in paraffin, and sectioned at 3 ⁇ m thickness.
  • Representative tumor sections from vehicle control and S-equol-treated mice are tested for Ki-67 expression to assess cell proliferation, and for ER ⁇ pY36.
  • Statistical significance in the experiments is assessed by two-tailed Student's t test. In all assays, p ⁇ 0.05 is considered statistically significant.
  • Ki67 is encoded by the gene MKI67 gene and is required to maintain individual mitotic chromosomes dispersed in the cytoplasm following nuclear envelope disassembly. Measuring the fraction of Ki-67-positive cells in a tumor is known to be a reliable parameter for assessing cancer patient prognosis. Subjects with TNBC are known to have very high levels of Ki67.
  • IHC Immunohistochemistry
  • Ki67 proliferation marker
  • ER ⁇ total ER ⁇
  • pY36-ER ⁇ phosphorylated ER ⁇
  • B7H1 also known as PDL-1
  • MIA and S 100B are conducted using core needle biopsy tissue samples prior to treatment.
  • Patients are given oral S-equol 50 mg, twice daily for approximately two weeks (10-21 days) prior to the scheduled oncologic surgeries or start of primary systemic therapy.
  • Tumor samples are obtained from the surgery or the two-week repeat biopsy.
  • Post-treatment IHC evaluation of Ki67, total ER ⁇ , phosphorylated ER ⁇ (pY36) and MIA and S 100B are obtained.
  • Ki67 reduction is compared pre- and post-treatment with S-equol.
  • S-equol causes a measurable decrease in Ki67, indicating its efficacy in this tumor type.
  • a c-DNA microarray platform is also used to discover downstream transcriptional targets of ER ⁇ resulting from activation by S-equol.
  • Patients eligible for the study have newly diagnosed, previously untreated, melanoma. All stages of the disease are eligible. They take S-equol during the time they undergo standard preoperative evaluation or pre-therapeutic work-up for advanced disease, such as staging procedures or central line placement. If primary surgery is performed, tissue leftover after standard diagnostic evaluation is used for the two-week biomarker assessment. If the patient does not have primary surgery at the end of the two-week S-equol treatment, a second set of biopsies is obtained before the start of any standard systemic therapy. See, FIG. 10 .
  • a standard procedure for acquiring core biopsy is utilized for determining melanoma prior to enrollment. Whenever possible, the tumor tissues used for IHC analyses are acquired at the same time as the diagnostic core biopsy. The tissue samples are sent for staining of total and pY36, ER ⁇ and Ki67, and analyzed.
  • Whole-Genome DASL HT platform from Illumina which is designed for gene expression profiling with extremely low input of RNA (50 pg), is used to interrogate paired FFPE tissue obtained before and after exposure to S-equol.
  • Statistical analysis is conducted which shows that ER ⁇ -expressing melanoma responds to S-equol as manifested by measurable decline in Ki67.
  • the primary analysis estimates the geometric mean change of the Ki67 expression from baseline to two weeks. This is performed using a one-sample t-test of the pre/post differences in the log-transformed data, and summarized by the 95% confidence interval. All computations are performed with SAS v9.2+ (Cary, N.C.) or R v2.15+ (Vienna, Austria).
  • a follow-up clinical study enrolls newly diagnosed melanoma patients, and patients are treated and assessed as discussed in Example 7, except that the dose of S-equol is increased to 150 mg twice daily. Treatment with S-equol is for 10-21 days. Ki67 is analyzed as described in Example 7.

Abstract

The present invention provides methods and compositions for treating or preventing melanoma with S-equol. The S-equol may be administered alone or in combination with one or more cytotoxic or immunotherapeutic compound or molecule.

Description

    CROSS-REFERENCE TO RELATED APPLICATIONS
  • This application claims priority from U.S. Provisional Application Ser. No. 62/685,392, filed Jun. 15, 2018, and U.S. Provisional Application Ser. No. 62/728,981, filed Sep. 10, 2018, which are hereby incorporated by reference in their entireties.
  • STATEMENT OF GOVERNMENTAL SUPPORT
  • This invention was made with U.S. Government support under grant number CA206529 awarded by the National Institutes of Health and the Texas Cancer Research Grant Contract under grant number DP150055 by the Cancer Prevention and Research Institute of Texas. The U.S. Government and the Texas State Government have certain rights in this invention.
  • BACKGROUND OF THE INVENTION Field of the Invention
  • This invention relates to methods and compositions for treating or preventing melanoma with a pharmaceutically effective amount of S-equol or a pharmaceutical composition comprising S-equol. In a preferred embodiment, the method relates to a combination therapy using S-equol and another anti-cancer treatment, such as immunotherapy.
  • Description of the Related Art
  • The methods and compositions of the present invention are suitable for various types of cancers, particularly those responsive to targeting of the estrogen receptor beta (ERβ). In addition to being suitable for treating breast cancer (see, U.S. Provisional Application 62/685,392, filed Jun. 15, 2018), the methods and compositions of the present invention are suitable for treating melanoma.
  • Malignant melanoma is a potentially serious skin cancer that results from the uncontrolled growth of pigment cells, called melanocytes. Melanoma may be the result of UV-induced changes to the skin, and its occurrence may be increased as a result of various genetic mutations. Mutations arising in the c-Kit gene, the BRAF gene, the CDKN2A gene, which regulates cell division, the MDm2 gene, which is a negative regulator of the p53 tumor suppressor gene, the retinoblastoma RB1 gene, and the MC1R (melanocortin-1-receptor) gene are among those that may increase the risk of developing melanoma. Treatment of melanoma may include the use of surgery, chemotherapy and/or immunotherapy. (Gupta et al., 2016; Clark et al., 2016; Steinberg et al., 2017).
  • Recent years have witnessed major clinical breakthroughs in cancer immunotherapies (Mellman et al. (2011); Kaufman et al. (2013); Brower (2015); Topalian et al. (2015)), which include blocking the immune-suppressive immune checkpoint molecules CTLA-4 (cytotoxic T-lymphocyte-associated protein 4 or CD152 [cluster of differentiation 152]), PD-1 (programmed cell death protein 1), and PD-L1 (programmed death ligand 1; Hodi et al. (2010); Wolchok et al. (2013); Topalian et al. (2014)). PD-1 is a checkpoint protein on T-cells that serves as an “off switch” to keep T cells from attacking other cells in the body. PD-1 interacts with PD-L1 on tumor cells. When PD-L1 interacts with PD-1, it prevents the T-cells from attacking the tumor cells. PD-1 inhibitors are currently being used to treat melanoma and other cancers.
  • A review of cancer immunotherapy using checkpoint blockade has been published as in Ribas and Wolchok, (2018) (see reference list herein), and is incorporated by reference herein in its entirety, particularly for the purposes of discussing pathways to be targeted with immunotherapy and antibodies for use in that checkpoint blockade. In addition, methods of rationally selecting cancer vaccine targets based on a patient's “mutanome,” namely, a set of somatic mutations that generate cancer-specific neoepitopes which can be recognized by autologous T cells as foreign, are discussed in Sahin and Tureci, (2018) (see reference list herein). This publication is also incorporated by reference herein in its entirety for discussing personalized targeting of tumor antigens such that cells of the immune system (e.g., CD4+ T helper cells and CD8+ T cytotoxic cells) can be activated to attack tumor cells. See, FIGS. 1 and 2. Both of these types of cancer immunotherapy can be combined with the use of S-equol in accordance with the present invention.
  • CD4+ T cells play a key role in the functioning of a healthy immune system. They assist B cells to make antibodies, activate the microbe killing capacity of macrophages and recruit other immune cells to infected or inflamed areas of the body. These activities are orchestrated through their production of various cytokines and chemokines. It has been known for some time that uncommitted CD4+ T-cells can differentiate into Th1 or Th2 cells, based on the prevailing pro-inflammatory/anti-inflammatory environment, and that these activated Th1 and Th2 cells had distinct cytokine production patterns and functions. Generally, Th1 cells were associated with the eradication of intracellular pathogens whereas Th2 cells were heavily involved in responses against extracellular pathogens and parasites. Uncontrolled Th1 responses were implicated in autoimmunity and aberrant Th2 responses were associated with allergy and asthma development. However, this model did not explain the observation that a deficiency in Th1 signaling and/or cytokines still allowed the development of autoimmune diseases such as rheumatoid arthritis and multiple sclerosis. More recently (2006) a third subset of CD4 T cells, Th17 cells, which have a pro-inflammatory bias was identified. Subsequent research using animal models and human studies has demonstrated a key role for Th17 cells in the immune system's defense against extracellular bacteria and fungi as well as the development of autoimmune diseases, mediated by the secretion of IL-17 by these cells. The secretion of IL-23 from antigen-presenting cells such as dendritic cells, which have been activated by the uptake and processing of pathogens, in turn activates Th17 cells. (Taken from Bitesized Immunology: https://www.immunology.org/public-information/bitesized-immunology/cells/th17-cells.) See, FIG. 2.
  • CD8+ (cytotoxic) T cells, like CD4+ Helper T cells, are generated in the thymus and express the T cell receptor. However, rather than the CD4 molecule, cytotoxic T cells express a dimeric coreceptor, CD8, usually composed of one CD8a and one CD8P chain. CD8+ T cells recognize peptides presented by MHC Class I molecules, found on all nucleated cells. The CD8 heterodimer binds to a conserved portion (the α3 region) of MHC Class I during T cell/antigen presenting cell interactions (see FIG. 1). CD8+ T cells (often called cytotoxic T lymphocytes, or CTLs) are very important for immune defense against intracellular pathogens, including viruses and bacteria, and for tumor surveillance. When a CD8+ T cell recognizes its antigen and becomes activated, it has three major mechanisms to kill infected or malignant cells. The first is secretion of cytokines, primarily TNF-α and IFN-γ, which have anti-tumor and anti-microbial effects. The second major function is the production and release of cytotoxic granules. These granules, also found in natural killer (NK) cells, contain two families of proteins, perforin, and granzymes. Perforin forms a pore in the membrane of the target cell, similar to the membrane attack complex of complement. This pore allows the granzymes also contained in the cytotoxic granules to enter the infected or malignant cell. Granzymes are serine proteases which cleave the proteins inside the cell, shutting down the production of viral proteins and ultimately resulting in apoptosis of the target cell. The cytotoxic granules are released only in the direction of the target cell, aligned along the immune synapse, to avoid non-specific bystander damage to healthy surrounding tissue (see FIG. 1). CD8+ T cells are able to release their granules, kill an infected cell, then move to a new target and kill again, often referred to as serial killing. The third major function of CD8+ T cell destruction of infected cells is via Fas/FasL interactions. Activated CD8+ T cells express FasL on the cell surface, which binds to its receptor, Fas, on the surface of the target cell. This binding causes the Fas molecules on the surface of the target cell to trimerize, which pulls together signaling molecules. These signaling molecules result in the activation of the caspase cascade, which also results in apoptosis of the target cell. Because CD8+ T cells can express both molecules, Fas/FasL interactions are a mechanism by which CD8+ T cells can kill each other, called fratricide, to eliminate immune effector cells during the contraction phase at the end of an immune response. In addition to their critical role in immune defense against viruses, intracellular bacteria, and tumors, CD8+ T cells can also contribute to an excessive immune response that leads to immunopathology, or immune-mediated damage. (Taken from Bitesized Immunology: https://www.immunology.org/public-information/bitesized-immunology/cells/cd8-t-cells.)
  • With respect to NK cells, the PK136 monoclonal antibody reacts with mouse NK1.1, an antigen expressed by natural killer cells and a subset of T cells in the NK1.1 mouse strains including C57BL and NZB. Several commonly used laboratory mouse strains such as BALB/c, SJL, AKR, CBA, C3H and A do not express the NK1.1 antigen. For detection of NK cells in these strains the monoclonal antibody DXS 14-5971 is used. Simultaneous staining of C57BL/6 spleen cells with PK136 and DXS reveals coexpression of both markers by a majority of cells as well as presence of small populations of DXS+PK136− and DXS−PK136+ cells.
  • CD45 (lymphocyte common antigen) and its associated molecules have also been shown to be important for T cell activation. CD45 is a receptor-linked protein tyrosine phosphatase that is expressed on all leucocytes, and which plays a crucial role in the function of these cells. On T cells the extracellular domain of CD45 is expressed in several different isoforms, and the particular isoform(s) expressed depends on the particular subpopulation of cell, their state of maturation, and whether or not they have previously been exposed to antigen. It has been established that the expression of CD45 is essential for the activation of T cells via the TCR, and that different CD45 isoforms display a different ability to support T cell activation. Although the tyrosine phosphatase activity of the intracellular region of CD45 has been shown to be crucial for supporting signal transduction from the TCR, the nature of the ligands for the different isoforms of CD45 have been elusive. Moreover, the precise mechanism by which potential ligands may regulate CD45 function is unclear. Interestingly, in T cells CD45 has been shown to associate with numerous molecules, both membrane associated and intracellular; these include components of the TCR-CD3 complex and CD4/CD8. In addition, CD45 is reported to associate with several intracellular protein tyrosine kinases including R561ck and p59fyn of the src family, and ZAP-70 of the Syk family, and with numerous proteins of 29-34 kDa. These CD45-associated molecules may play an important role in regulating CD45 tyrosine phosphatase activity and function. However, although the role of some of the CD45-associated molecules (e.g. CD45-AP and LPAP) has become better understood in recent years, the role of others still remains obscure. See, Altin and Sloan, (1997) in the reference list herein.
  • However, these highly promising immunotherapies are only effective for a subset of cancer patients and are not usually curative. In particular, clinical trials of anti-PD-1 antibodies in triple negative breast cancer (TNBC) patients demonstrate only modest efficacy, with objective responses in the range of 10-20%, and an additional 20% of patients experiencing some stabilization of disease that would otherwise be rapidly progressive (Nanda et al. (2016); Emens et al. (2015); Dirix et al. (2015); Rugo et al. (2015); Dawood and Rugo (2016)). There is thus a pressing clinical need to identify better therapies to improve individual responses to immunotherapy for the treatment of TNBC, melanoma, and other cancers.
  • PD-1 and its ligand PD-L1 are immune checkpoint molecules that dampen T cell immunity (Lin et al. (2010); Curiel et al. (2003); Brahmer et al. (2012); Pardoll and Drake (2012); Pardoll (2012)). PD-L1 is overexpressed in tumor cells and some immune cells, including Tregs (Lin et al. (2010); Curiel et al. (2003); Pardoll (2012); Topalian et al. 2012)). Immunotherapy with anti-PD-1 (αPD-1) and anti-PD-LI antibodies (αPD-L1) has proven successful for treating various cancers (Brahmer (2012); Pardoll and Drake (2012); Pardoll (2012)). Early clinical trials for TNBC indicate that PD-1 and PD-L1 are valid targets for intervention and toxicities of treatment are mild. In a Phase 1 study of 27 women with heavily pretreated, chemotherapy resistant, metastatic TNBC that expressed PD-L1, the response rate was 18.4% to pembrolizumab, an αPD-1 antibody (Nanda et al. 2016). A similar study of atezolizumab, an αPD-LI antibody, produced a response rate of 19% in 21 evaluable, PD-L1 expressing TNBC tumors (Emens et al. (2015); Dawood and Rugo (2016)). Another group reported an 8.6% response rate to avelumab, another αPD-LI antibody, in 168 TNBC tumors unselected for PD-L1 expression (Dirix et al. 2015), indicating that level of PD-L 1 tumor expression may be important in mediating tumor response. Some disease stabilization was seen in this aggressive subtype of breast cancer, in approximately 20% of patients in these 3 trials. Serious toxicities were few and related to immune modulation. These clinical data indicate that the PD-1/PD-L1 axis can be a therapeutic target, likely in combination approaches for better efficacy.
  • There are two distinct types of estrogen receptors that mediate diverse physiological effects of estrogens on breast cancer cells. The first receptor is the ERα receptor which supports estrogen-dependent breast tumor growth. The second is the ERβ receptor which appears to work in an opposite fashion to significantly attenuate the growth of breast tumor cells in preclinical models (Clarke et al. (2003); Deroo and Korach (2006); Deroo and Buensuceso (2010); Honma et al (2008); Katzenellenbogen and Katzenellenbogen (2000); McDonnell and Norris (2002); Murphy and Watson (2006); Shaaban et al (2008). Thus, ERβ can be viewed in as a tumor suppressor gene in breast cancer. ERβ is expressed in about 40% of TNBC cancers, and the therapeutic potentials of targeting ERβ have not yet been investigated in TNBC. Two recent advances have now made it feasible to move from the preclinical models and into clinical research. The first advance is a newly discovered mechanism of rallying ERβ's antitumor activity (Harris (2007); Hartman et al (2009); Heldring et al. (2007); Thomas and Gustafsson (2011)), and the second is the development of an oral formulation of an ERβ agonist (S-equol) that has already been tested for clinical safety, pharmacodynamics and tolerance in humans (Jackson et al. (2011a & b); Setchell et al. (20005); Setchell (2002); Schwen et al. (2012).
  • Estrogen receptor (ERβ; nuclear or cytoplasmic) is reported to be present in approximately half of TNBC (Marotti et al. (2010); Reese et al. (2014)) as well as in melanoma (Marzagalli et al., 2016). In addition to its tumor-intrinsic activity, ERβ in host cells has also been implicated in melanoma tumor inhibition (Cho et al. (2010)). In support of a tumor-extrinsic antitumor activity of ERβ, Cho et al. showed that syngeneic murine melanoma cells grafted to recipient ERβ-KO animals grew more robustly than those in WT recipient. In this study, the authors showed that the growth of 3 different skin cancer cell lines in their syngeneic murine hosts was sensitive to the activity of the estrogen receptor. Therefore these transplantation studies, used as models of skin cancer induction by SSUV irradiation, strongly suggest that signaling by the ERβ is an endogenous protective mechanism to inhibit skin cancer growth that is partially mediated immunologically. The recognition of selective ER modulators (SERMs) as agents able to elicit estrogenic effects in a tissue-specific manner has expanded the potential population that could benefit from ER ligand therapies. (Smith and O'Malley, 2018).
  • Thus, rallying ERβ antitumor activity through ERβ-specific modification and/or ligand binding represents an excellent therapeutic opportunity for TNBC and melanoma. However, the therapeutic potentials of targeting ERβ have not been extensively exploited, partly due to the paucity in the knowledge of how to harness its antitumor activity. A phosphotyrosine residue (Y36) in ERβ, but not ERα, has recently been identified that is important for regulating the antitumor activity of ERβ in TNBC cells (Yuan et al. (2014); Yuan et al. (2016)).
  • When this phosphotyrosine residue (pY36) was purposefully mutated by adding a phenylalanine group (Y36F), activation of ERβ target genes were decreased in a TNBC cell line (MDA-MB-231). The Y36F mutation also obliterated the ability of ERβ to inhibit tumor cell growth in vitro and in vivo. This preclinical research therefore strongly supports the importance of pY36 in the antitumor activity of ERβ (Yuan et al., 2014).
  • There is also evidence that pY36 status correlates with survival of breast cancer patients. Research has been done using a Prognostic Tissue Microarray (TMA) from the National Cancer Institute (NCI), which consists of a large cohort of breast tumor samples with a clinical follow-up record. Using a total of 726 readable IHC samples, patients with pY36-negative tumors were found to have statistically significant shorter disease-free and overall survival than those with pY36-positive tumors. Interestingly, the association with survival was only seen in Stage II & III disease, which raises the intriguing possibility that pY36 activity may have an effect on disease progression from locally advanced to metastatic breast cancer. Collectively, pY36 intensity appears to have a stronger correlation with patient outcome than total ERβ, underscoring the clinical importance of this previously unappreciated phosphotyrosine switch. See, FIG. 3.
  • S-equol, an ERβ agonist, was previously shown to increase respiratory and maximal glycolysis fluxes in rat hippocampal neurons, as well as cytochrome oxidase (COX) activity and COX1 protein levels in brains from ovariectomized mice, and has been studied in human subjects to assess its health impact and safety (Yao et al. (2013); Jenks et al. (2002); Jackson et al. (2011a & b); Usui et al., (2013)).
  • S-equol can be produced either chemically (i.e., chemical synthesis) or by biotransformation (biosynthesis) through the metabolism of daidzein, an isoflavone found in soy and red clover, by gut bacteria. The structure of S-equol is shown below.
  • Figure US20190380998A1-20191219-C00001
  • Equol has a chiral center and therefore can exist in two enantiomeric forms. S-equol, R-equol, racemic equol, and non-racemic mixtures of equol (collectively “equol”); compositions of equol; anhydrous crystalline polymorph of equol; processes for the preparation of equol; and methods of using equol are described in U.S. Pat. No. 8,716,497 (filed Sep. 10, 2012); U.S. Pat. No. 8,048,913 (filed Sep. 14, 2009); U.S. Pat. No. 7,960,432 (filed Jul. 3, 2008); U.S. Pat. No. 7,396,855 (filed Jul. 24, 2003); U.S. Pat. No. 8,263,790 (filed Jun. 1, 2011); U.S. Pat. No. 7,960,573 (filed May 4, 2009); U.S. Pat. No. 7,528,267 (filed Aug. 1, 2005); U.S. Pat. No. 8,668,914 (filed Jul. 31, 2009); U.S. Pat. No. 8,580,846 (filed Aug. 18, 2006); U.S. Pat. No. 8,450,364 (filed Apr. 9, 2012); and U.S. Pat. No. 8,153,684 (filed Oct. 2, 2009); U.S. Pat. No. 9,408,824 (filed Mar. 5, 2014); and U.S. Pat. No. 9,914,718 (filed Oct. 14, 2015); each of which is hereby incorporated by reference in its entirety.
  • Formulations comprising isoflavones and products derived therefrom have been used in the past to treat disease. For example, a mixture of equol, genistein, and daidzein, or a mixture of equol, genistein, daidzein, and IBSO03569 have shown potential for treating or preventing neurodegeneration and Alzheimer's disease. See Zhao et al. (2009); U.S. Pat. No. 8,552,057; Yao et al. (2013) (collectively “Brinton et al.”). S-equol alone has also been described for treating Alzheimer's Disease. See, U.S. application Ser. No. 15/659,114 published as U.S. Patent Publication 2018/0028491, and International Patent Publication WO/2018/022604, which are hereby incorporated by reference in their entireties.
  • However, there remains a need in the art for methods that utilize S-equol for the treatment of other disease states that have in the past been difficult to treat. In particular, there is a need in the art for methods that utilize S-equol in the treatment of melanoma.
  • SUMMARY OF THE INVENTION
  • The following brief summary is not intended to include all features and aspects of the present invention, nor does it imply that the invention must include all features and aspects discussed in this summary.
  • The inventors have found that S-equol, preferably pure and isolated S-equol, benefit melanoma patients. In particular, the inventors have found that S-equol in combination with immunotherapy is particularly effective in treating melanoma.
  • It is therefore an object of the invention to provide the following:
      • 1. A method for treating or preventing melanoma by administering a pharmaceutically effective amount of a formulation comprising S-equol to a subject in need thereof.
      • 2. The method of Item 1, wherein the formulation comprises 10-200 mg S-equol.
      • 3. The method of Item 1, wherein the formulation comprises 50-150 mg S-equol.
      • 4. The method of Item 1, wherein the formulation comprises about 50 mg S-equol.
      • 5. The method of Item 1, wherein the formulation comprises about 150 mg S-equol.
      • 6. The method of Item 1, wherein the formulation is administered orally, intravenously, intraperitoneally, or subcutaneously.
      • 7. The method of Item 1, wherein said subject is a human.
      • 8. The method of Item 1, wherein the S-equol is administered in combination with one or more other cancer treatments.
      • 9. The method of Item 8, wherein the S-equol is administered in combination with an immunotherapeutic agent.
      • 10. The method of Item 9, wherein the immunotherapeutic agent is an antibody.
      • 11. The method of Item 10, wherein the antibody is directed to programmed cell death protein 1 (PD-1).
      • 12. The method of Item 11, wherein the antibody is directed to programmed death ligand 1 (PDL-1).
      • 13. The method of Item 11, wherein the antibody is pembrolizumab.
      • 14. The method of Item 12, wherein the antibody is atezolizumab.
      • 15. The method of Item 12, wherein the antibody is avelumab.
      • 16. The method of Item 1, wherein the formulation is essentially free of genistein, daidzein, and/or IBSO03569.
      • 17. The method of Item 1, wherein genistein, daidzein, and/or IBSO03569 are not co-administered with S-equol.
      • 18. The method of Item 1, wherein the formulation is essentially free of R-equol.
      • 19. The method of Item 1, wherein the S-equol is produced chemically.
      • 20. The method of Item 1, wherein the formulation is administered once per day.
      • 21. The method of Item 1, wherein the formulation is administered twice per day.
      • 22. The method of Item 1, wherein the formulation is administered three times per day.
      • 23. The method of Item 1, wherein the formulation is administered four times per day.
  • The invention also relates to compositions comprising S-equol as described for the methods herein.
  • The foregoing and other objects, features and advantages of the invention will be apparent from the following more particular description of preferred embodiments of the invention, as illustrated in the accompanying drawings in which like reference characters refer to the same parts throughout the different views.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1. Schematic depiction of the action of CD8+ cells.
  • FIG. 2. Schematic depiction of the action of CD4+ cells.
  • FIG. 3. Association of pY36 with disease outcomes in breast cancer. FIG. 3A shows the Kaplan-Meier estimate of disease-free and overall survival in Stages II-III specimens correlating with IHC intensity of pY36-ERβ. FIG. 3B shows the Kaplan-Meier estimate of disease-free and overall survival in Stages II-III specimens correlating with IHC intensity of (A) and total ERβ IHC.
  • FIG. 4. A schema for combination therapy. Mice were administered one of: (1) vehicle+rat IgG2a; (2) S-equol+rat IgG2a; (3) vehicle+anti-PD-1 antibody; or (4) S-equol+anti-PD-1 antibody. Mice receiving anti-PD-1 antibody were given 250 μg every 3 days, intraperitoneally, and mice receiving S-equol were given 50 mg/kg per day by oral gavage. Treatment started 7 days after tumor challenge with B16 melanoma.
  • FIG. 5. A role of ERβ signaling in immune cells. FIG. 5A shows the schema for the chimera experiments. FIG. 5B shows tumor growth in chimeric mice (WT>WT: n=10, KI>WT: n=4). p=0.0003. FIG. 5C shows total CD8+ T cells from tumors. FIG. 5D shows interferon gamma (IFN-γ) expressing CD8+ T cells from tumors.
  • FIG. 6. Effect of S-equol in combination with an anti-PD-1 antibody on mice challenged with an B16 melanoma tumor cell line over 12 days. Mice were administered anti-PD-1 antibody at 250 μg every 3 days, intraperitoneally, and S-equol at 50 mg/kg per day by oral gavage. FIG. 6 shows that separately, S-equol and anti-PD-1 antibody reduce tumor volume compared to control, but S-equol in combination with an anti-PD-1 antibody reduces tumor volume compared to S-equol or anti-PD-1 antibody alone over 12 days post-challenge.
  • FIG. 7. Effect of S-equol in combination with an anti-PD-1 antibody on mice challenged with an B16 melanoma tumor cell line over 19 days. Mice were administered anti-PD-1 antibody at 250 μg every 3 days, intraperitoneally, and S-equol at 50 mg/kg per day by oral gavage. FIG. 7 shows that separately, S-equol and anti-PD-1 antibody reduce tumor volume compared to control, but S-equol in combination with an anti-PD-1 antibody reduces tumor volume compared to S-equol or anti-PD-1 antibody alone over 19 days post-challenge.
  • FIG. 8. Effect of S-equol in combination with an anti-PD-1 antibody on tumor weight in mice challenged with an B16 melanoma tumor cell line. FIG. 8A shows that separately, S-equol and anti-PD-1 antibody reduce tumor weight compared to control, but S-equol in combination with an anti-PD-1 antibody reduces tumor weight compared to S-equol or anti-PD-1 antibody alone. FIG. 8B shows mouse total body weight for each treatment.
  • FIG. 9. Effect of S-equol in combination with an anti-PD-1 antibody and LY500307 on mice challenged with an B16 melanoma tumor cell line over 16 days. Mice were administered anti-PD-1 antibody at 250 μg every 3 days, intraperitoneally, and S-equol at 50 mg/kg per day by oral gavage. FIG. 9A shows that separately, S-equol and anti-PD-1 antibody reduce tumor volume compared to control, but S-equol in combination with an anti-PD-1 antibody reduces tumor volume compared to S-equol or anti-PD-1 antibody alone over 12 days post-challenge. FIG. 9B shows results with LY500307, a selective ERβ agonist (SERBA-1; Erteberel®).
  • FIG. 10. Generalized protocol for human clinical trial with S-equol.
  • DETAILED DESCRIPTION OF THE INVENTION
  • Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by those of ordinary skill in the art to which this invention belongs. Although any methods and materials similar or equivalent to those described herein can be used in the practice or testing of the present invention, the preferred methods and materials are described. Generally, nomenclatures utilized in connection with, and techniques of, cell and molecular biology and chemistry are those well-known and commonly used in the art. Certain experimental techniques, not specifically defined, are generally performed according to conventional methods well known in the art and as described in various general and more specific references that are cited and discussed throughout the present specification. For purposes of clarity, the following terms are defined below.
  • “Substantially pure” means about 90% pure, preferably 95% pure, and more preferably 98% pure of any contaminating proteins.
  • “Substantially free” means about less than about 10%, preferably less than 5%, and more preferably less than 2% of any contaminating proteins.
  • “Approximately” or “about” means within +/−10%, preferably within +/−5%, more preferably within +/−2% of that which is being measured.
  • The present invention relates to the prevention and/or treatment of melanoma with S-equol.
  • Dosage amounts and administration schedules for S-equol will depend on whether the S-equol is being administered prophylactically to a patient at risk of developing melanoma, or being administered as treatment for a patient already diagnosed with melanoma. A person at risk for developing melanoma may be a person with a family history of melanoma, and/or may have one or more mutations in a CDKN2 A gene, MDm2 gene, RB1 gene, or MC1R gene. In a patient already diagnosed with melanoma, dosages and administration schedules may also vary depending on the stage of the cancer (stage I, stage II, stage III, stage IV or stage V), the number of lymph nodes involved, tumor size and the availability of and/or decision to co-administer other therapies. Dosages and administration schedules may also vary depending on various molecular markers present on or in tumor cells, including, but not limited to Hsp90, RGS1, osteopontin, HER3, ING4, ING3, NCOA3, MCM4, MCM6, Ki67 and pY36. Diagnostic tests for markers which are prognostic for the aggressiveness and/or likelihood of recurrence of a tumor may be used as a factor or determining dosage regimens. (Gogas et al. (2009), incorporated by reference in its entirety for all purposes.)
  • S-equol can be administered one or more times per day at 1-400 mg per dose, more preferably 10-320 mg, more preferably 50-150 mg. Non-limiting examples include 2 mg, 5 mg, 10 mg, 15 mg, 20 mg, 40 mg, 50 mg, 80 mg, 100 mg, 150 mg, 160 mg, 200 mg, 250 mg, 300 mg 320 mg, etc. or approximately or about those doses. The dose may be administered one, two, three or four times per day, preferably twice per day (B.I.D.) The regimen can be continued indefinitely, or for such time in intervals where markers are examined and responsiveness to the dose is determined. Examples of such intervals are two weeks, four weeks, two months, four months, six months, etc. or about or approximately those intervals. No upper limit, with respect to administration schedule, is required.
  • The S-equol administered is preferably formulated for oral administration; however, other routes of administration are also contemplated, including rectal, optical, buccal (for example sublingual), parenteral (for example subcutaneous, intramuscular, intradermal and intravenous) and transdermal administration.
  • Compositions or formulations according to the present invention can comprise one or more pharmaceutically-acceptable or industrial standard fillers. The filler must not be deleterious to a subject treated with the composition. The filler can be solid or a liquid, or both. The filler can be formulated with the active S-equol as a unit-dose, for example a tablet, which can typically contain from about 10% to 80% by weight of S-equol. Compositions can be prepared by any of the well known techniques of pharmacy, for example admixing the components, optionally including excipients, diluents (for example water) and auxiliaries as are well known in the pharmaceutical field.
  • Compositions suitable for oral administration can be presented in discrete units, such as capsules, cachets, lozenges, or tablets, each containing a predetermined amount of the extract; as a powder or granules; as a solution or a suspension in an aqueous or non-aqueous liquid; or as an oil-in-water or water-in-oil emulsion. Such compositions can be prepared by any suitable method of pharmacy which includes the step of bringing into association the active S-equol and one or more suitable carriers (which can contain one or more accessory ingredients as noted above). In general the compositions of the invention are prepared by uniformly and intimately admixing the S-equol with a liquid or finely divided solid carrier, or both, and then, if necessary, shaping the resulting mixture. For example, a tablet can be prepared by comprising or moulding a powder or granules containing the extract, optionally with one or more accessory ingredients. Compressed tablets can be prepared by compressing in a suitable machine, the extracts in the form of a powder or granules optionally mixed with a binder, lubricant, inert diluents, and/or surface active/dispersing agent(s). Moulded tablets can be made by moulding, in a suitable machine, the powdered compound moistened with an inert liquid binder.
  • Suitable fillers, such as sugars, for example lactose, saccharose, mannitol or sorbitol, cellulose preparations and/or calcium phosphates, for example tricalcium phosphate or calcium hydrogen phosphate, and also binders such as starch pastes using, for example, corn, wheat, rice or potato starch, gelatin, tragacanth, methylceullose and/or polyvinylpyrrolidone, and, if desired, disintegrators, such as the above-mentioned starches, also carboxymethyl starch, cross linked polyvinyl pyrrolidone, agar or alginic acid or a salt thereof, such as sodium alginate. Excipients can be flow conditioners and lubricants, for example silicic acid, talc, stearic acid or salts thereof, such as magnesium or calcium stearate, and/or polyethylene glycol. Dragee cores are provided with suitable, optionally enteric, coatings, there being used, inter alia, concentrated sugar solutions which can comprise gum arabic, talc, polyvinylpyrrolidone, polyethylene glycol and/or titanium dioxide, or coating solutions in suitable organic solvents or solvent mixtures, or, for the preparation of enteric coatings, solutions of suitable cellulose preparations, such as microcrystalline cellulose, acetylcellulose phthalate or hydroxypropylmethylcellulose phthalate. Dyes or pigments can be added to the tablets or dragee coatings, for example for identification purposes or to indicate different doses of active ingredients.
  • Other orally administrable pharmaceutical compositions are dry-filled capsules made, for example, of gelatin, and soft, sealed capsules made of gelatin and a plasticiser, such as glycerol or sorbitol. The dry-filled capsules can comprise the extracts in the form of granules, for example in admixture with fillers, such as lactose, binders, such as starches, and/or glicants, such as talc or magnesium stearate, and, where appropriate, stabilizers. In soft capsules, the extract is preferably dissolved or suspended in suitable liquids, such as fatty oils, paraffin oil or liquid polyethylene glycols, to which stabilizers can also be added.
  • According to one aspect of the invention, the compositions comprising S-equol include those described in U.S. Pat. No. 7,960,432 (filed Jul. 3, 2008); U.S. Pat. No. 7,396,855 (filed Jul. 24, 2003); and U.S. Pat. No. 9,408,824 (filed Mar. 5, 2014)—the disclosures of each are hereby incorporated by reference in their entireties.
  • According to another aspect of the invention, S-equol can be prepared chemically (i.e., chemical synthesis) according to the processes described in U.S. Pat. No. 8,716,497 (filed Sep. 10, 2012); U.S. Pat. No. 8,263,790 (filed Jun. 1, 2011); U.S. Pat. No. 7,960,573 (filed May 4, 2009); U.S. Pat. No. 7,528,267 (filed Aug. 1, 2005) and U.S. Pat. No. 9,914,718 (filed Oct. 14, 2014)—the disclosures of each are hereby incorporated by reference in their entireties. For example, S-equol can be enantioselectively prepared using an iridium catalyst with a chiral ligand. These methods of enantioselectively preparing S-equol are incorporated by reference.
  • According to another aspect of the invention, S-equol can be a single anhydrous crystalline polymorph of S-equol, such as the anhydrous crystalline polymorph of S-equol described in U.S. Pat. No. 9,914,718 (application Ser. No. 14/883,617, filed Oct. 14, 2015)—the disclosure of which, including the chemical and physical properties used to characterize the anhydrous crystalline polymorph of S-equol, is incorporated by reference in their entireties. For example, the anhydrous crystalline polymorph of S-equol described in U.S. Patent Application Publication No. 2016/0102070 has the following characteristic X-ray powder diffraction pattern wavenumbers (cm−1): 3433, 3023, 3003, 2908, 2844, 1889, 1614, 1594, 1517, 1508, 1469, 1454, 1438, 1400, 1361, 1323, 1295, 1276, 1261, 1234, 1213, 1176, 1156, 1116, 1064, 1020, 935, 897, 865, 840, 825, 810, 769, 734, 631, 616, 547, 517, 480, and 461. The characterizations of anhydrous crystalline polymorph of S-equol are incorporated by reference.
  • The S-equol can be administered in combination with one or more additional cancer treatments, including surgery, cytotoxic chemotherapy, radiation, immunotherapy, cancer vaccines, inhibitors of cellular pathways (protein or peptide, nucleic acid-based [antisense oligonucleotides including DNA oligonucleotides, antisense siRNA, shRNA] and/or hormonal (adjuvant endocrine) therapy. Examples of cytotoxic chemotherapy suitable for treating melanoma include, but are not limited to imiquimod cream (Zyclara®), dacarbazine (DTIC) and temozolomide (Temodar®). Examples of immunotherapy include but are not limited to nivolumab (Opdivo®), pembrolizumab (Keytruda®), nivolumab (Opdivo®), and ipilirurab (Yervoy®), imatinib (Glevac®), nilotinib (Tasigna®) or interferon and interleukin-2 (IL-2). Other options include injections of the T-VEC vaccine (Imlygic®), or the Bacille Calmette-Guerin (BCG) vaccine.
  • Appropriate dosages of immunotherapy monoclonal antibodies or other cancer therapeutic are determined based on the indication, and their determination is well within the skill in the art. Dosing may be in mg, in mg/kg or mg/m2. See, Sachs et al. (2016), which is incorporated by reference herein in its entirety for all purposes, including examples of therapeutic antibodies, small molecules and dosages. The S-equol and/or additional therapeutic composition may be administered once, twice, three times or more per day, depending on the dose, toxicity and indication. Dosing may occur over several days, weeks or months, and may be continuous or intermittent. Typical dosages for immunotherapeutics are 50-500 mg/day, more preferably 100-400 mg/day, more preferably 150-250 mg/day. In certain embodiments, the dose may be 150 mg/day or 150 mg twice per day, or 250 mg/day or 250 mg twice a day. In other embodiments the dosage may be given based on patient weight, for instance 0.5-10 mg/kg, more preferably about 1, 2 or 5 mg/kg. Dosage may also be given as mg/m2, for example 50-500 mg/m2, more preferably 100-300 mg/m2, for example about 100, 150, 200, 250 or 300 mg/m2.
  • The dosages may be about or greater than the lower end of any of the afore-stated ranges, or about or less than the upper end of any of the afore-stated ranges.
  • Responsiveness to the S-equol+/−other cancer therapy can be assessed by analyzing one or more cellular markers, by imaging techniques, by cellular proliferation assays and by direct examination of tumors. The analysis of cellular markers can be done with immunohistochemical or immunocytochemical techniques or by nucleic acid detection, such as hybridization techniques and polymerase chain reaction (PCR). A preferred biomarker to assess mitotic division and tumor growth is Ki67 (Beelen et al. (2012); Urruticoechea et al. (2005)).
  • The following examples are provided to aid the understanding of the present invention, the true scope of which is set forth in the appended claims. It is understood that modifications can be made in the procedures set forth without departing from the spirit of the invention.
  • EXAMPLES
  • The processes of the present invention will be better understood in connection with the following examples, which are intended as an illustration only and without limiting the scope of the invention. Various changes and modifications to the disclosed embodiments will be apparent to those skilled in the art and such changes and modifications including, without limitation, those relating to the processes, formulations and/or methods of the invention may be made without departing from the spirit of the invention and the scope of the appended claims.
  • Example 1
  • To determine whether the newly identified phosphotyrosine switch regulates ERβ tumor-extrinsic function, a whole-body knock-in (K1) mouse model (C57BL/6) was established in which the corresponding tyrosine residue of endogenous mouse ERβ is mutated to phenylalanine (Y-F). Consistent with previously reported ERβ knockout (KO) mice (Krege et al. (1998)), these phosphorylation mutant KI mice had no overt developmental defects and were grossly indistinguishable from their WT littermates (unpublished data). Survey of ERβ expression in KO and KI mice indicated that the Y-F mutant protein was expressed at levels comparable to WT ERβ in multiple tissues, including bone marrow and spleen (FIG. 4A). Syngeneic murine tumor cells of various origins were then transplanted, including the MMTV-Wnt mammary tumor cell line with a basal-like breast cancer profile (Pfefferle et al. (2013)), into WT and KI recipient mice. In all cases, tumor cells grew more robustly in ERβ KI recipient mice than in their syngeneic WT counterparts (FIG. 4B, and data not shown). These data clearly demonstrate that the phosphotyrosine switch is important for ERβ tumor-extrinsic antitumor activity in multiple tumor types.
  • Example 2
  • The following experiment sought to delineate further the host ERβ signaling in tumor inhibition based on the role of antitumor immunity and recent clinical advances in cancer immunotherapies (Topalian et al. (2015); Chen and Flies (2013); Sharma and Allison (2015)). A mouse chimera experiment was conducted involving a bone marrow transplant. As illustrated in FIG. 5A, WT recipient mice were first irradiated (10 Gy) to kill endogenous bone marrow cells, followed by transplant with bone marrow from syngeneic KI or WT donors. After confirming successful chimerism in KI>WT and WT>WT mice, tumor cells were injected 8 weeks after bone marrow transplant. Tumor growth was significantly greater in KI>WT chimeras (with KI immune cells) versus WT>WT controls (FIG. 5B). This suggests that KI immune cells poorly controlled tumor growth. Thus, these data link the importance of tumor extrinsic ERβ antitumor activity to the immune response.
  • Example 3
  • Next, tumor-infiltrating immune cell populations were analyzed. Total numbers of tumor-infiltrating CD4+ and CD8+ T cells were reduced in KI>WT versus WT>WT mice (FIG. 5C and data not shown), further supporting the notion that antitumor immunity in KI mice is compromised. Furthermore, the prevalence of IFNγ-producing CD8+ (antitumor) cells was significantly lower in tumors from KI>WT versus WT>WT mice (FIG. 5D), suggesting compromised cytotoxic potency of CD8+ T cells in the absence of functional ERβ signaling. Additional preliminary data not shown here indicate that activation of dendritic cells, which prime antitumor T cells, was also compromised in KI>WT chimeric mice, as evidenced by their reduced MHC-II expression. Furthermore, effector T cells were less activated (lower CD44/CD62L) and had other reduced effector functions in KI>WT chimeras [e.g., lower tumor necrosis factor alpha (TNF)α, interleukin (IL)-2, and perforin, data not shown]. These data strongly suggest ERβ-dependent augmentation of antitumor CD8+ T cell effector activity and improved intra-tumor immune cell accumulation. Therefore, rallying ERβ antitumor activity with clinically safe ERβ agonists such as S-equol will improve efficacy of existing anticancer immunotherapies and make them more effective to treat melanoma.
  • Example 4
  • Melanoma cell line B16 was tested to determine the effects of a PD-1 inhibitor, S-equol and a combination of the two therapies on tumor growth. This experimental design includes four arms: (1) controls, (2) αPD-1, (3) S-equol, and (4) αPD-1+S-equol (FIG. 4) by injecting into mice 5×105 cells subcutaneously. Antibodies were administered α-PD-1: 250 μg/mice by i.p. injection every 3 days and S-equol was administered at 50 mg/kg/day by oral gavage. Treatment began 7 day after tumor challenge. Tumor growth trajectories were compared within treated mice using a repeated measures linear mixed model. The primary outcome was the logarithm of the tumor size, and the test statistic is the treatment X time interaction. Tumors were measured about 10 times. The number of recipient mice use used in the studies (n=6 per group) was based on the above study. Results (FIGS. 6-9) from this experiment demonstrate the ability of S-equol to boost anticancer immunotherapy.
  • Example 5
  • In addition to measurements of tumor size and weight in each arm, immunophenotyping is performed to gain more mechanistic insight into the antitumor effects of mono- and combinational therapies. Tumors, spleens, and draining lymph nodes are harvested and weighed. Anti-coagulated blood was collected by cardiac puncture. Tumors are paraffin-embedded for immunohistochemistry and snap-frozen for mRNA and protein analysis by Luminex. Flow cytometry is used (for phenotype/functions) and ViCell (for quantification) to analyze immune cells from tumor infiltrates, tumor-draining lymph nodes, and spleens. Analyses include CD3+ total T cells and CD4+ and CD8+ T cell subsets; effector function (e.g., IFN-γ, TNF-α, IL-2, perforin, CD107a); activation (e.g., CD69, CD44, CD62L); and exhaustion (e.g., PD-1, Tim3, Lag3). Antigen-presenting cells (CD11b+CD1c monocyte/macrophages, CD11b+CD11c+ dendritic cells), NK1.1+NKp46+ NK cells, regulatory T cells (Treg, CD3+CD4+CD25hiFoxp3+), and myeloid-derived suppressor cells (MDSC, CD11b+Gr−1hi) are also analyzed. CD8+ T cell receptor diversity were assessed with a commercial kit (Adaptive Biotechnologies). Local cell proliferation is also tested with Ki67 and BrdU stain.
  • Example 6
  • Further work in a preclinical animal model supports the notion of using S-equol to treat melanoma. In immunocompromised mouse experiments, using B16, a murine tumor cell line know to be useful to study human melanoma metastasis and solid tumor formation, growth of tumors is suppressed with S-equol treatment compared to control. 0.5×106 B16 cells are injected into normal BL mice. When the tumor masses reach 50 to 80 mm3 (about one week after the inoculation), the mice are given daily subcutaneous injections of S-equol (20 or 60 mg/kg per day) or PBS as a vehicle control. Tumor development is followed by caliper measurements along two orthogonal axes: length (L) and width (W) and volume (V) is estimated by the formula V=[L×(W2)]/2. Tumors harvested from mice are fixed in 10% neutral-buffered formalin, dehydrated, embedded in paraffin, and sectioned at 3 μm thickness. Representative tumor sections from vehicle control and S-equol-treated mice are tested for Ki-67 expression to assess cell proliferation, and for ERβ pY36. Statistical significance in the experiments is assessed by two-tailed Student's t test. In all assays, p<0.05 is considered statistically significant.
  • Example 7
  • A pilot biomarker clinical study is performed in 20 subjects with documented melanoma to determine the effect of the ERβ agonist S-equol on Ki67 an indicator of tumor cell proliferation. Tumor cell proliferation is measured by immunohistological staining for Ki-67. Ki-67 is encoded by the gene MKI67 gene and is required to maintain individual mitotic chromosomes dispersed in the cytoplasm following nuclear envelope disassembly. Measuring the fraction of Ki-67-positive cells in a tumor is known to be a reliable parameter for assessing cancer patient prognosis. Subjects with TNBC are known to have very high levels of Ki67.
  • Patients enrolled in the study receive a history, physical, laboratory assessment, radiological imaging, and diagnostic biopsies. Immunohistochemistry (IHC) of Ki67 (proliferation marker), total ERβ, and pY36-ERβ (phosphorylated ERβ), B7H1 (also known as PDL-1) and MIA and S 100B are conducted using core needle biopsy tissue samples prior to treatment. Patients are given oral S-equol 50 mg, twice daily for approximately two weeks (10-21 days) prior to the scheduled oncologic surgeries or start of primary systemic therapy. Tumor samples are obtained from the surgery or the two-week repeat biopsy. Post-treatment IHC evaluation of Ki67, total ERβ, phosphorylated ERβ (pY36) and MIA and S 100B are obtained. Ki67 reduction is compared pre- and post-treatment with S-equol. S-equol causes a measurable decrease in Ki67, indicating its efficacy in this tumor type. A c-DNA microarray platform is also used to discover downstream transcriptional targets of ERβ resulting from activation by S-equol.
  • Patients eligible for the study have newly diagnosed, previously untreated, melanoma. All stages of the disease are eligible. They take S-equol during the time they undergo standard preoperative evaluation or pre-therapeutic work-up for advanced disease, such as staging procedures or central line placement. If primary surgery is performed, tissue leftover after standard diagnostic evaluation is used for the two-week biomarker assessment. If the patient does not have primary surgery at the end of the two-week S-equol treatment, a second set of biopsies is obtained before the start of any standard systemic therapy. See, FIG. 10.
  • A standard procedure for acquiring core biopsy is utilized for determining melanoma prior to enrollment. Whenever possible, the tumor tissues used for IHC analyses are acquired at the same time as the diagnostic core biopsy. The tissue samples are sent for staining of total and pY36, ERβ and Ki67, and analyzed. Whole-Genome DASL HT platform from Illumina, which is designed for gene expression profiling with extremely low input of RNA (50 pg), is used to interrogate paired FFPE tissue obtained before and after exposure to S-equol.
  • Statistical analysis is conducted which shows that ERβ-expressing melanoma responds to S-equol as manifested by measurable decline in Ki67. The primary analysis estimates the geometric mean change of the Ki67 expression from baseline to two weeks. This is performed using a one-sample t-test of the pre/post differences in the log-transformed data, and summarized by the 95% confidence interval. All computations are performed with SAS v9.2+ (Cary, N.C.) or R v2.15+ (Vienna, Austria).
  • Example 8
  • A follow-up clinical study enrolls newly diagnosed melanoma patients, and patients are treated and assessed as discussed in Example 7, except that the dose of S-equol is increased to 150 mg twice daily. Treatment with S-equol is for 10-21 days. Ki67 is analyzed as described in Example 7.
  • REFERENCES
  • The patent and scientific literature referred to herein establishes the knowledge that is available to those with skill in the art. All United States patents and published or unpublished United States patent applications cited herein are incorporated by reference. All published foreign patents and patent applications cited herein are hereby incorporated by reference. All other published references, documents, manuscripts and scientific literature cited herein are hereby incorporated by reference.
    • Altin, J. G. and Sloan, E. K. The role of CD45 and CD45-associated molecules in T cell activation. Immunol. Cell Biol. 75(5):430-435 (1997).
    • Baum M., Buzdar A. U., Cuzick J., et al. Anastrozole alone or in combination with tamoxifen versus tamoxifen alone for adjuvant treatment of postmenopausal women with early breast cancer: first results of the ATAC randomized trial. Lancet. 359: 2131-2139, (2002).
    • Beelen K., Zwart W., Linn S. C. Can predictive biomarkers in breast cancer guide adjuvant endocrine therapy? Nat Rev Clin Oncol. 9: 529-541, (2012).
    • Brahmer, J. R. et al. Safety and activity of anti-PD-L 1 antibody in patients with advanced cancer. N Engl J Med 366, 2455-2465, doi:10.1056/NEJMoal200694 (2012).
    • Brower, V. Checkpoint blockade immunotherapy for cancer comes of age. J Natl Cancer Inst 107, doi: 10.1093/jnci/djv069 (2015).
    • Chen, L. & Flies, D. B. Molecular mechanisms of T cell co-stimulation and co-inhibition. Nat Rev Immunol 13, 227-242, doi:10.1038/nri3405 (2013).
    • Chia, K. & Tutt, A. Triple-negative breast cancer: An update. Adv Breast Cancer 4, 75-80, (2007).
    • Cho, J. L., Allanson, M. & Reeve, V. E. Oestrogen receptor-beta signalling protects against transplanted skin tumour growth in the mouse. Photochem Photobiol Sci 9, 608-614, doi: 10.1039/b9pp00168a (2010).
    • Clark C A, Gupta H B, Sareddy G, Pandeswara S, Lao S, Yuan B, Drerup J M, Padron A, Conejo-Garcia J, Murthy K, Liu Y, Turk M J, Thedieck K, Hurez V, Li R, Vadlamudi R, Curiel T J. Tumor-Intrinsic P D-L1 Signals Regulate Cell Growth, Pathogenesis, and Autophagy in Ovarian Cancer and Melanoma,” Cancer Res. 2016 Dec. 1; 76(23):6964-6974.
    • Clarke R., Liu M., Bouker K. B., et al. Antiestrogen resistance in breast cancer and the role of estrogen receptor signaling. Oncogene. 22: 7316-7339, (2003).
    • Cleator, S., Heller, W. & Coombes, R. C. Triple-negative breast cancer: therapeutic options. Lancet Oncol. 8, 235-244, (2007).
    • Coates et al. “Tailoring therapies—improving the management of early breast cancer: St. Gallen International Expert Consensus on the Primary Therapy of Early Breast Cancer 2015,” Annals of Oncology 26(8):1533-46, (2015)
    • Curiel, T. J. et al. Blockade of B7-H1 improves myeloid dendritic cell-mediated antitumor immunity. Nat Med 9, 562-567, doi: 10.1038/nm863 (2003).
    • Davies C., Pan H., Godwin J., et al. Long-term effects of continuing adjuvant tamoxifen to 10 years versus stopping at 5 years after diagnosis of estrogen receptor-positive breast cancer: ATLAS, a randomized trial. Lancet. 381: 805-816, 2013.
    • Dawood, S. & Rugo, H. S. Targeting the host immune system: PD-1 and PD-L 1 antibodies and breast cancer. Curr Opin Support Palliat Care 10, 336-342, doi:10.1097/SPC.0000000000000243 (2016).
    • Dent, R. et al. Triple-negative breast cancer: clinical features and patterns of recurrence. Clin Cancer Res. 13, 4429-4434 (2007).
    • Deroo B. J., Korach K. S. Estrogen receptors and human disease. J Clin Invest. 116: 561-570, (2006).
    • Deroo B. J., Buensuceso A. V. Minireview: Estrogen receptor-beta: mechanistic insights from recent studies. Mol Endocrinol. 24: 1703-1714, (2010).
    • Diaz, L. K., Cryns, V. L., Symmans, W. F. & Sneige, N. Triple negative breast carcinoma and the basal phenotype: From expression profiling to clinical practice. Advances in Anatomic Pathology 14, 419-430 (2007).
    • Dirix, L. Y., Takacs, I. & Nikolinakos, P. e. a. Avelumab (MSB0010718C), an anti-PD-LI antibody, in patients with locally advanced or metastatic breast cancer: A phase 1b JA YELIN solid tumor trial. 2015 San Antonio Breast Cancer Symposium Abstract S1-04, (2015).
    • Emens, L. A., Braiteh, F. S. & Cassier, P. e. a. Inhibition of PD-L 1 by MPDL3280A leads to clinical activity in patients with metastatic triple-negative breast cancer (TNBC). The American Association for Cancer Research Annual Meeting Abstract 2859, (2015).
    • Gerson, R., Alban, F., Villalobos, A. & Serrano, A. Recurrence and survival rates in early breast cancer cases with triple negative immunophenotype. Gae Med Mex 144, 27-34, (2008).
    • Gonzalez-Angulo, A. M. Advances in triple receptor-negative breast cancer. Clin Adv Hematol Oncol 5, 956-957 (2007).
    • Goss P. E., Ingle J. N., Martino S., et al. Randomized trial of letrozole following tamoxifen as extended adjuvant therapy in receptor-positive breast cancer: updated findings from NCIC CTG MA.17. J Natl Cancer Inst. 97: 1262-1271, (2005).
    • Gupta, H. B. et al. “Tumor cell-intrinsic PD-L1 promotes tumor-initiating cell generation and functions in melanoma and ovarian cancer,” Signal Transduction and Targeted Therapy (2016) 1, 16030.
    • Haffty, B. G. et al. Locoregional relapse and distant metastasis in conservatively managed triple negative early-stage breast cancer. J Clin Oncol. 24, 5652-5657 (2006).
    • Harris H. A. Estrogen receptor-beta: recent lessons from in vivo studies. Mol Endocrinol. 21: 1-13, (2007).
    • Hartman J., Strom A., Gustafssom J. A. Estrogen receptor beta in breast cancer—diagnostic and therapeutic implications. Steroids. 74: 635-641, (2009).
    • Heldring N., Pike A., Anderrson S., et al. Estrogen receptors: how do they signal and what are their targets. Physiol Rev. 87: 905-931, (2007).
    • Hodi, F. S. et al. Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med 363, 711-723, doi: 10.1056/NEJMoal003466 (2010).
    • Honma N., Hori R., Iwase T. et al: Clinical importance of estrogen receptor-beta evaluation in breast cancer patients treated with adjuvant tamoxifen therapy. J Clin Oncol. 26: 3727-3734, (2008).
    • Irvin, W. J. J. & Carey, L. A. What is triple-negative breast cancer? Eur J Cancer. 44, 2799-2805, (2008).
    • Jackson R. L., Greiwe J. S., Schwen R. J. Emerging evidence of the health benefits of S-equol, an estrogen receptor beta agonist. Nutr Rev. 69: 432-448, (2011a).
    • Jackson R. L., Greiwe J. S., Schwen R. J. Single-dose and steady-state pharmacokinetic studies of S-equol, a potent nonhormonal, estrogen receptor beta-agonist being developed for the treatment of menopausal symptoms. Menopause, 18 185-193, (2011b)
    • Jenks et al., A pilot study on the effects of S-equol compared to soy isoflavones on menopausal hot flash frequency, Journal of Women's Health, 21 (2012) 674-682, (2002).
    • Kang, S. P., Martel, M. & Harris, L. N. Triple negative breast cancer: current understanding of biology and treatment options. Curr Opin Obstet Gynecol. 20, 40-46, (2008).
    • Kaplan, H. G. & Malmgren, J. A. Impact of triple negative phenotype on breast cancer prognosis. Breast Journal 14, 456-463, (2008).
    • Katzenellenbogen B., Katzenellenbogen J. Estrogen receptor transcription and transactivation: Estrogen receptor alpha and estrogen receptor beta: regulation by selective estrogen receptor modulators and importance in breast cancer. Breast Cancer Res. 2: 335-344, (2000).
    • Kaufman, H. L. et al. The Society for Immunotherapy of cancer consensus statement on tumour immunotherapy for the treatment of cutaneous melanoma. Nat Rev Clin Oncol 10, 588-598, doi: 10.1038/nrclinonc.2013.153 (2013).
    • Kittaneh et al. “Molecular profiling for breast cancer: A comprehensive review,” Biomark Cancer 5:61-70, (2013).
    • Krege, J. H. et al. Generation and reproductive phenotypes of mice lacking estrogen receptor beta. Proc Natl Acad Sci USA 95, 15677-15682, (1998).
    • Lin, P. Y. et al. B7-H1-dependent sex-related differences in tumor immunity and immunotherapy responses. J Immunol 185, 2747-2753, doi:10.4049/jimmunol. 1000496 (2010).
    • Marotti, J. D., Collins, L. C., Hu, R. & Tamimi, R. M. Estrogen receptor-beta expression in invasive breast cancer in relation to molecular phenotype: results from the Nurses' Health Study. Modern Pathology 23, 197-204, doi:10.1038/modpathol.2009. 158, (2010).
    • Marzagalli M., et al. Estrogen Receptor P in Melanoma: From Molecular Insights to Potential Clinical Utility, Frontiers in Endocrinology, Vol. 7, Article 140 (2016).
    • McDonnell D., Norris J.: Connections and regulation of the human estrogen receptor. Science. 296: 1642-1644, (2002).
    • Mellman, I., Coukos, G. & Dranoff, G. Cancer immunotherapy comes of age. Nature 480, 480-489, doi:10.1038/nature10673 (2011).
    • Murphy L. C., Watson P. H. Is oestrogen receptor-beta a predictor of endocrine therapy responsiveness in human breast cancer? Endocr Relat Cancer. 13: 327-334, (2006).
    • Nanda, R. et al. Pembrolizumab in patients with advanced triple-negative breast cancer: Phase 1b KEYNOTE-012 Study. J Clin Oncol 34, 2460-2467, doi:10.1200/JC0.2015.64.8931 (2016).
    • Padrón Á, Hurez V, Gupta H B, Clark C A, Pandeswara S L, Yuan B, Svatek R S, Turk M J, Drerup J M, Li R, Curiel T J. “Age effects of distinct immune checkpoint blockade treatments in a mouse melanoma model,” Exp Gerontol. 2018 May; 105:146-154.
    • Pardoll, D. & Drake, C. Immunotherapy earns its spot in the ranks of cancer therapy. J Exp Med 209, 201-209, doi:10.1084/jem.20112275 (2012).
    • Pardoll, D. M. The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer 12, 252-264, doi: 10.1038/nrc3239 (2012).
    • Pfefferle, A. D. et al. Transcriptomic classification of genetically engineered mouse models of breast cancer identifies human subtype counterparts. Genome Biol 14, R125, doi:10.1186/gb-2013-14-11-r125 (2013).
    • Reese, J. M. et al. ERβ: characterization, prognosis, and evaluation of treatment strategies in ER alpha positive and -negative breast cancer. BMC Cancer 14, 749, doi:10.1186/1471-2407-14-749 (2014).
    • Reis-Filho, J. S. & Tutt, A. N. J. Triple negative tumours: A critical review. Histopathology 52, 108-118, (2008).
    • Ribas, A. and Wolchok, J. D. Cancer immunotherapy using checkpoint blockade. Science 359:1350-1355, (2018).
    • Rugo, H. S. et al. Preliminary efficacy and safety of pembrolizumab (MK-3475) in patients with PD-LI-positive, estrogen receptor-positive (ER+)/HER2-negative advanced breast cancer enrolled in KEYNOTE-028. 2015 San Antonio Breast Cancer Symposium Abstract S5-07 (2015).
    • Sachs, J. R. et al. Optimal dosing for targeted therapies in oncology: drug development cases leading by example. Clin. Cancer Res. 22(6):OF1-7 (2016).
    • Sahin, U. and Tureci, O. Personalized vaccines for cancer immunotherapy. Science 359:1355-1356, (2018).
    • Santen R. J., Brodie H., Simpson E. R., et al. History of aromatase: saga of an important biological mediator and therapeutic target. Endocr Rev. 30: 343-375, (2009).
    • Schwen, R. J., Greiwe, J. S., Schwen, R. J. Elucidation of the metabolic pathway of s-equol in rat, monkey and man. Food Chemistry Toxicology. 50: 2074-2083, (2012).
    • Setchell, K. D., Clerici C., Lephart E. D., et al. S-equol, a potent ligand for estrogen receptor beta, is the exclusive enantiomeric form of the soy isoflavone metabolite produced by human intestinal bacterial flora. Am J Clini Nutr. 81: 1072-1079, (2005).
    • Setchell, K. D. The clinical importance of the metabolite equol—a clue to the effectiveness of soy and its isoflavones. Nature. 132:3577-3584, (2002).
    • Shaaban A. M., Green A. R., Karthik S. Nuclear and cytoplasmic expression of ERbeta1, ERbeta2, and ERbeta5 identifies distinct prognostic outcome for breast cancer patients. Clin Cancer Res. 14: 5228-5235, (2008).
    • Sharma, P. & Allison, J. P. Immune checkpoint targeting in cancer therapy: toward combination strategies with curative potential. Cell 161, 205-214, doi:10.1016/j.cell.2015.03.030 (2015).
    • Shou J., Massarweh S., Osborne C. K., et al. Mechanisms of tamoxifen resistance: increased estrogen receptor-HER2/neu cross-talk in ER/HER2-positive breast cancer. J Natl Cancer Inst. 96: 926-935, 2004.
    • Smith I. E., Dowsett M., Ebbs S., et al. Neoadjuvant treatment of postmenopausal breast cancer with anastrozole, tamoxifen, or both in combination: The immediate preoperative anastrozole, tamoxifen, or combined with tamoxifen (IMPACT) multicenter double-blind randomized trial. J Clini Onc. 23: 5108-5116, (2005).
    • Smith C. L., O'Malley B. W. Coregulator Function: A Key to Understanding Tissue Specificity of Selective Receptor Modulators. Endocrine Reviews 25(1):45-71, (2004).
    • Steinberg S M, Shabaneh T B, Zhang P, Martyanov V, Li Z, Malik B T, Wood T A, Boni A, Molodtsov A, Angeles C V, Curiel T J, Whitfield M L, Turk M J. “Myeloid Cells That Impair Immunotherapy Are Restored in Melanomas with Acquired Resistance to BRAF Inhibitors,” Cancer Res. 2017 Apr. 1; 77(7):1599-1610.
    • Thomas C., Gustafsson J. A. The different roles of ER subtypes in cancer biology and therapy. Nat Rev Cancer. 11: 597-608, (2011).
    • Topalian, S. L., Drake, C. G. & Pardoll, D. M. Immune checkpoint blockade: a common denominator approach to cancer therapy. Cancer Cel 27, 450-461, doi:10.1016/j.ccell.2015.03.001 (2015).
    • Topalian, S. L. et al. Survival, durable tumor remission, and long-term safety in patients with advanced melanoma receiving nivolumab. J Clin Oncol 32, 1020-1030, doi:10.1200/JC0.2013.53.0105 (2014).
    • Topalian, S. L., Drake, C. G. & Pardall, D. M. Targeting the PD-1/B7-H1(PD-L1) pathway to activate anti-tumor immunity. Curr Opin Immunol 24, 207-212, doi:10.1016/j.coi.2011.12.009 (2012).
    • Urruticoechea A., Smith I., Dowsett M. Proliferation marker Ki-67 in early breast cancer. Journal of Clinical Oncology 23:7212-7220, (2005).
    • Usui et al., Effects of natural S-equol supplements on overweight or obesity and metabolic syndrome in the Japanese, based on sex and equol status, Clinical endocrinology, 78 365-372, (2013).
    • Wolchok, J. D. et al. Nivolumab plus ipilimumab in advanced melanoma. N Engl J Med 369, 122-133, doi:10.1056/NEJMoa1302369 (2013).
    • Yao et al., Potentiation of brain mitochondrial function by S-equol and R/S-equol estrogen receptor beta-selective phytoSERM treatments, Brain Research, 1514 128-141, (2013).
    • Yuan, B. et al. A phosphotyrosine switch determines the antitumor activity of ERbeta. J Clin Invest 124, 3378-3390, doi:10.1172/JC174085 (2014).
    • Yuan B., Cheng L., Chiang H. C., et al. Mobilizing ERβ antitumor activity through a phosphotyrosine switch. J Clini Invest. 124(8):3378-90, (2014).
    • Yuan, B. et al. Tyrosine phosphorylation regulates ERbeta ubiquitination, protein turnover, and inhibition of breast cancer. Oncotarget, doi: 10.18632/oncotarget. 10018 (2016).
    • Zhao et al., A select combination of clinically relevant phytoestrogens enhances estrogen receptor β-binding selectivity and neuroprotective activities in vitro and in vivo. Neuroendocrinology, 150(2):770-783, (2009).

Claims (23)

1. A method for treating or preventing melanoma, comprising administering a pharmaceutically effective amount of a formulation comprising S-equol to a subject in need thereof.
2. The method of claim 1, wherein the formulation comprises 10-200 mg S-equol.
3. The method of claim 1, wherein the formulation comprises 50-150 mg S-equol.
4. The method of claim 1, wherein the formulation comprises about 50 mg S-equol.
5. The method of claim 1, wherein the formulation comprises about 150 mg S-equol.
6. The method of claim 1, wherein the formulation is administered orally, intravenously, intraperitoneally, or subcutaneously.
7. The method of claim 1, wherein said subject is a human.
8. The method of claim 1, wherein the S-equol is administered in combination with one or more other cancer treatments.
9. The method of claim 8, wherein the S-equol is administered in combination with an immunotherapeutic agent.
10. The method of claim 9, wherein the immunotherapeutic agent is an antibody.
11. The method of claim 10, wherein the antibody is directed to programmed cell death protein 1 (PD-1).
12. The method of claim 11, wherein the antibody is directed to programmed death ligand 1 (PDL-1).
13. The method of claim 11, wherein the antibody is pembrolizumab.
14. The method of claim 12, wherein the antibody is atezolizumab.
15. The method of claim 12, wherein the antibody is avelumab.
16. The method of claim 1, wherein the formulation is essentially free of genistein, daidzein, and/or IBSO03569.
17. The method of claim 1, wherein genistein, daidzein, and/or IBSO03569 are not co-administered with S-equol.
18. The method of claim 1, wherein the formulation is essentially free of R-equol.
19. The method of claim 1, wherein the S-equol is produced chemically.
20. The method of claim 1, wherein the formulation is administered once per day.
21. The method of claim 1, wherein the formulation is administered twice per day.
22. The method of claim 1, wherein the formulation is administered three times per day.
23. The method of claim 1, wherein the formulation is administered four times per day.
US16/384,428 2018-06-15 2019-04-15 Methods of treating and preventing melanoma with s-equol Abandoned US20190380998A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US16/384,428 US20190380998A1 (en) 2018-06-15 2019-04-15 Methods of treating and preventing melanoma with s-equol
US17/819,863 US20220387380A1 (en) 2018-06-15 2022-08-15 Methods of treating and preventing melanoma with s-equol

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US201862685392P 2018-06-15 2018-06-15
US201862728981P 2018-09-10 2018-09-10
US16/384,428 US20190380998A1 (en) 2018-06-15 2019-04-15 Methods of treating and preventing melanoma with s-equol

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US17/819,863 Continuation US20220387380A1 (en) 2018-06-15 2022-08-15 Methods of treating and preventing melanoma with s-equol

Publications (1)

Publication Number Publication Date
US20190380998A1 true US20190380998A1 (en) 2019-12-19

Family

ID=66397460

Family Applications (2)

Application Number Title Priority Date Filing Date
US16/384,428 Abandoned US20190380998A1 (en) 2018-06-15 2019-04-15 Methods of treating and preventing melanoma with s-equol
US17/819,863 Pending US20220387380A1 (en) 2018-06-15 2022-08-15 Methods of treating and preventing melanoma with s-equol

Family Applications After (1)

Application Number Title Priority Date Filing Date
US17/819,863 Pending US20220387380A1 (en) 2018-06-15 2022-08-15 Methods of treating and preventing melanoma with s-equol

Country Status (5)

Country Link
US (2) US20190380998A1 (en)
EP (1) EP3843718A1 (en)
JP (1) JP2021535187A (en)
AU (1) AU2019285641A1 (en)
WO (1) WO2019240872A1 (en)

Families Citing this family (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP4005567A1 (en) 2016-04-06 2022-06-01 Noxopharm Limited Isoflavonoid composition with improved pharmacokinetics
US11229703B2 (en) 2016-04-06 2022-01-25 Noxopharm Limited Radiotherapy improvements
JP7329860B2 (en) * 2018-06-15 2023-08-21 ボード オブ レジェンツ,ザ ユニバーシティ オブ テキサス システム Method for treating and preventing breast cancer using S-equol
MX2021015418A (en) * 2019-07-17 2022-04-12 Noxopharm Ltd Immuno-oncology therapy using isoflavone compounds.
JP2023519990A (en) 2020-03-30 2023-05-15 ノクソファーム リミティド Methods for treating inflammation associated with infection
US20220160678A1 (en) * 2020-11-24 2022-05-26 Board Of Regents, The University Of Texas System Modulation of molecular markers using s-equol

Family Cites Families (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU2003259220C1 (en) 2002-07-24 2017-08-24 Australian Health & Nutrition Association Limited Compositions and products containing enantiomeric equol, and methods for their making
WO2004039327A2 (en) 2002-10-29 2004-05-13 Colorado State University Research Foundation Use of equol for treating androgen mediated diseases
US8668914B2 (en) 2002-07-24 2014-03-11 Brigham Young University Use of equol for treating skin diseases
US20050245492A1 (en) * 2004-04-28 2005-11-03 Lephart Edwin D Use of equol for treating skin diseases
US8580846B2 (en) 2002-10-29 2013-11-12 Brigham Young University Use of equol for ameliorating or preventing neuropsychiatric and neurodegenerative diseases or disorders
US7528267B2 (en) 2005-08-01 2009-05-05 Girindus America, Inc. Method for enantioselective hydrogenation of chromenes
US20080108696A1 (en) 2006-08-02 2008-05-08 Brinton Roberta D Phytoestrogenic Formulations for Alleviation or Prevention of Neurodegenerative Diseases
JP2013126374A (en) * 2010-03-04 2013-06-27 Kyushu Univ Gene related to action of isoflavones
CN105828834A (en) * 2013-11-05 2016-08-03 同源生物服务股份有限公司 Combinations of checkpoint inhibitors and therapeutics to treat cancer
WO2016061184A1 (en) 2014-10-14 2016-04-21 Jackson Richard L Anhydrous crystalline form of s-equol
CA2980297A1 (en) * 2015-03-20 2016-09-29 Syndax Pharmaceuticals, Inc. Combination of hdac inhibitor and anti-pd-1 antibody for treatment of cancer
EA201792273A1 (en) * 2015-04-17 2018-04-30 Бристол-Маерс Сквибб Компани COMPOSITIONS CONTAINING A COMBINATION OF ANTI-PD-1 ANTIBODIES AND OTHER ANTIBODIES
US10391079B2 (en) 2016-07-26 2019-08-27 Ausio Pharmaceuticals, Llc Methods of diagnosing and treating Alzheimer's disease with S-equol

Also Published As

Publication number Publication date
JP2021535187A (en) 2021-12-16
EP3843718A1 (en) 2021-07-07
WO2019240872A1 (en) 2019-12-19
US20220387380A1 (en) 2022-12-08
AU2019285641A1 (en) 2021-04-08

Similar Documents

Publication Publication Date Title
US20220387380A1 (en) Methods of treating and preventing melanoma with s-equol
US11090286B2 (en) Methods of treating and preventing breast cancer with S-equol
Akbay et al. Activation of the PD-1 pathway contributes to immune escape in EGFR-driven lung tumors
Mao et al. Selective blockade of B7‐H3 enhances antitumour immune activity by reducing immature myeloid cells in head and neck squamous cell carcinoma
Triebel et al. A soluble lymphocyte activation gene-3 (sLAG-3) protein as a prognostic factor in human breast cancer expressing estrogen or progesterone receptors
Márquez-Garbán et al. Antiestrogens in combination with immune checkpoint inhibitors in breast cancer immunotherapy
JP2021502071A (en) Cancer biomarkers and how to use them
Zhu et al. CD8+/FOXP3+ ratio and PD-L1 expression associated with survival in pT3N0M0 stage esophageal squamous cell cancer
WO2018148378A1 (en) Modulating biomarkers to increase tumor immunity and improve the efficiacy of cancer immunotherapy
US20200297728A1 (en) Methods of treating cancer
JP2021502342A (en) Adenosine pathway inhibitors for cancer treatment
Navarrete-Bernal et al. Biological landscape of triple negative breast cancers expressing CTLA-4
US20230113705A1 (en) Methods for diagnosing, prognosing and managing treatment of breast cancer
Li et al. PD‑L1/PD‑1 blockade in breast cancer: The immunotherapy era
Zhao et al. Neutrophils resist ferroptosis and promote breast cancer metastasis through aconitate decarboxylase 1
Slovin Biomarkers for immunotherapy in genitourinary malignancies
Zhou et al. Orphan nuclear receptor TLX promotes immunosuppression via its transcriptional activation of PD-L1 in glioma
TW201823471A (en) Methods of treating patients with a retinoic acid receptor-[alpha] agonist and an anti-CD38 antibody
US20220160678A1 (en) Modulation of molecular markers using s-equol
JP2015524252A (en) Methods for determining personalized treatment compositions for prostate cancer and breast cancer
Kam et al. ENOX2 inhibition enhances infiltration of effector memory T-cell and mediates response to chemotherapy in immune-quiescent nasopharyngeal carcinoma
KR20170071393A (en) Composition for diagnosis of radioresistance and use thereof
Xia et al. Integrated bioinformatic analysis and cell line experiments reveal the significant role of the novel immune checkpoint TIGIT in kidney renal clear cell carcinoma
Weng et al. Aging and biliary tract cancers: Epidemiology, molecular biology, and clinical practice
He et al. Myeloid-derived suppressor cells in head and neck squamous cell carcinoma

Legal Events

Date Code Title Description
STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

AS Assignment

Owner name: THE BOARD OF REGENTS OF THE UNIVERSITY OF TEXAS SYSTEM, TEXAS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:LI, RONG;YUAN, BIN;CURIEL, TYLER;SIGNING DATES FROM 20190115 TO 20190201;REEL/FRAME:055866/0129

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: FINAL REJECTION MAILED

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION