Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
METHODS AND CHEMICALS FOR TREATMENT OF MEK RELATED PATHOLOGIES
Document Type and Number:
WIPO Patent Application WO/2019/055493
Kind Code:
A1
Abstract:
A method for treating a mitogen activated protein kinase kinase/extracellular signal-regulated kinase ("MEK")-induced or an MEK-related pathology in a mammal, comprising administering a pharmaceutical composition containing an effective amount of a first therapeutic, wherein the first therapeutic includes a first MEK inhibitor, or a pharmaceutically acceptable salt, solvate, ester, amide, clathrate, stereoisomer, enantiomer, prodrug or analog thereof. According to one embodiment, the first MEK inhibitor is 3β-Hydroxy-5α, 25R-spirostan-12-(3'-methylphenyl thiosemicarbazone), the mammal is human, and the MEK-induced or an MEK-related pathology is breast cancer.

Inventors:
EL SAYED KHALID A (US)
ELSAYED HEBA E (EG)
Application Number:
PCT/US2018/050632
Publication Date:
March 21, 2019
Filing Date:
September 12, 2018
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
BOARD OF SUPERVISORS FOR THE UNIV OF LOUISIANA SYSTEM (US)
International Classes:
A61K31/175; A61K31/58; C07C337/08; C07J71/00
Domestic Patent References:
WO2016094904A12016-06-16
Foreign References:
US20070049591A12007-03-01
Other References:
LIAGRE ET AL.: "Inhibition of human rheumatoid arthritis synovial cell survival by hecogenin and tigogenin is associated with increased apoptosis, p38 mitogen-activated protein kinase activity and upregulation of cyclooxygenase-2", INTERNATIONAL JOURNAL OF MOLECULAR MEDICINE, vol. 20, 2007, pages 451 - 460, XP055583206
ELSAYED ET AL.: "Rationally designed hecogenin thiosemicarbazone analogs as novel MEK inhibitors for the control of breast malignancies", BIOORGANIC & MEDICINAL CHEMISTRY, vol. 25, no. 24, 27 September 2017 (2017-09-27), pages 6297 - 6312, XP085254613, DOI: 10.1016/j.bmc.2017.09.033
Attorney, Agent or Firm:
HOLOUBEK, Charles G. et al. (US)
Download PDF:
Claims:
CLAIMS

Wherefore, we claim:

1 . A method for treating a mitogen activated protein kinase kinase/extracellular signal-regulated kinase ("MEK")-induced or an MEK-related pathology in a mammal, comprising:

administering a pharmaceutical composition containing an effective amount of a first therapeutic;

wherein the first therapeutic includes a first MEK inhibitor, or a pharmaceutically acceptable salt, solvate, ester, amide, clathrate, stereoisomer, enantiomer, prodrug or analog thereof.

2. The method of claim 1 wherein the first MEK inhibitor is (3p,5a,25R)-3- Hydroxyspirostan-12-one.

3. The method of claim 1 wherein the first MEK inhibitor is a Hecogenin-12- thiosemicarbazones.

4. The method of claim 1 wherein the first MEK inhibitor is one of

3p-Hydroxy-5a,25R-spirostan-12-(phenyl thiosemicarbazone),

3p-Hydroxy-5a,25R-spirostan-12-(3'-chloroyphenyl thiosemicarbazone),

3p-Hydroxy-5a,25R-spirostan-12-(5'-chlorophenyl thiosemicarbazone),

3p-Hydroxy-5a,25R-spirostan-12-(3',5'-dichlorophenyl thiosemicarbazone), 3p-Hydroxy-5a,25R-spirostan-12-(3',7'-dichlorphenyl thiosemicarbazone), 3p-Hydroxy-5a,25R-spirostan-12-(5'-fluorophenyl thiosemicarbazone),

3p-Hydroxy-5a,25R-spirostan-12-(3'-nitrophenyl thiosemicarbazone),

3p-Hydroxy-5a,25R-spirostan-12-(5'-nitrophenyl thiosemicarbazone),

3p-Hydroxy-5a,25R-spirostan-12-(3'-trifluoromethylphenyl

thiosemicarbazone),

3p-Hydroxy-5a,25R-spirostan-12-(5'-trifluoromethylphenyl

thiosemicarbazone),

3p-Hydroxy-5a,25R-spirostan-12-(4',6'-di- triflouromethylphenylthiosemicarbazone),

3p-Hydroxy-5a,25R-spirostan-12-(3'-methoxyphenyl thiosemicarbazone), 3p-Hydroxy-5a,25R-spirostan-12-(5'-methoxyphenyl thiosemicarbazone), 3p-Hydroxy-5a,25R-spirostan-12-(5'-phenoxyphenyl thiosemicarbazone), 3p-Hydroxy-5a,25R-spirostan-12-(3'-ethylphenyl thiosemicarbazone),

3p-Hydroxy-5a,25R-spirostan-12-(3'-methylphenyl thiosemicarbazone), 3p-Hydroxy-5a,25R-spirostan-12-(5'-methylphenyl thiosemicarbazone), 3p-Hydroxy-5a,25R-spirostan-12-(3',5'-dimethylphenyl thiosemicarbazone), 3p-Hydroxy-5a,25R-spirostan-12-(3',7'-dimethylphenyl thiosemicarbazone), and

3p-Hydroxy-5a,25R-spirostan-12-(4',5'-methylenedioxyphenyl

thiosemicarbazone).

5. The method of claim 4 wherein the pharmaceutical composition contains a second therapeutic for the MEK-induced pathology or MEK-related pathology distinct from the first therapeutic.

6. The method of claim 5, wherein the second therapeutic is not a MEK inhibitor.

7. The method of claim 5, wherein the second therapeutic is one of second therapeutic could be one of Evista (Raloxifene Hydrochloride), Raloxifene Hydrochloride, Tamoxifen Citrate, Abemaciclib, Abraxane (Paclitaxel Albumin- stabilized Nanoparticle Formulation), Ado-Trastuzumab Emtansine, Afinitor (Everolimus), Anastrozole, Aredia (Pamidronate Disodium), Arimidex (Anastrozole), Aromasin (Exemestane), Capecitabine, Cyclophosphamide, Docetaxel, Doxorubicin Hydrochloride, Ellence (Epirubicin Hydrochloride), Epirubicin Hydrochloride, Eribulin Mesylate, Everolimus, Exemestane, 5-FU (Fluorouracil Injection), Fareston (Toremifene), Faslodex (Fulvestrant), Femara (Letrozole), Fluorouracil Injection, Fulvestrant, Gemcitabine Hydrochloride, Gemzar (Gemcitabine Hydrochloride), Goserelin Acetate, Halaven (Eribulin Mesylate), Herceptin (Trastuzumab), Ibrance (Palbociclib), Ixabepilone, Ixempra (Ixabepilone), Kadcyla (Ado-Trastuzumab Emtansine), Kisqali (Ribociclib), Lapatinib Ditosylate, Letrozole, Lynparza (Olaparib), Megestrol Acetate, Methotrexate, Neratinib Maleate, Nerlynx (Neratinib Maleate), Olaparib, Paclitaxel, Paclitaxel Albumin-stabilized Nanoparticle Formulation, Palbociclib, Pamidronate Disodium, Perjeta (Pertuzumab), Pertuzumab, Ribociclib, Tamoxifen Citrate, Taxol (Paclitaxel), Taxotere (Docetaxel), Thiotepa, Toremifene, Trastuzumab, Trexall (Methotrexate), Tykerb (Lapatinib Ditosylate), Verzenio (Abemaciclib), Vinblastine Sulfate, Xeloda (Capecitabine), Zoladex (Goserelin Acetate).

8. The method of claim 5, wherein the second therapeutic includes a second MEK inhibitor.

9. The method of claim 1 wherein the MEK-induced or MEK-related pathology is cancer or pre cancer state.

10. The method of claim 9 wherein the cancer is one of a skin cancer, breast cancer, prostate cancer, brain cancer, lung cancer, colorectal cancer, pancreatic cancer, ovarian cancer, liver cancer, bladder cancer, head and neck cancer, esophageal cancer, gastric cancer, and renal cancer.

1 1 . The method of claim 1 wherein the MEK-induced or MEK-related pathology is a non-cancer pathology.

12. The method of claim 1 1 wherein the non-cancer pathology is one of Alzheimer's disease, osteoarthritis, rheumatoid arthritis, hypertrophic cardiomyopathy, heart failure, stroke, septic shock, organ transplant rejection, and an RAS/RAF/MEK/ERK driven pathology.

13. The method of claim 12 wherein the RAS/RAF/MEK/ERK driven pathology includes one of Neurofibromatosis Type 1 , Cardiofaciocutaneous syndrome, Noonan syndrome, Noonan syndrome with multiple lentigines (LEOPARD syndrome), Costello syndrome, and Legius syndrome.

14. The method of claim 1 wherein

the mammal is human,

the MEK induced or MEK related pathology is a cancer,

the pharmaceutical composition contains a second therapeutic for the cancer; and

the first MEK inhibitor is a Hecogenin-12-thiosemicarbazones.

15. The method of claim 14 wherein the cancer is a breast cancer.

16. The method of claim 15 where the first MEK inhibitor is 3p-Hydroxy-5a, 25R- spirostan-12-(3'-methylphenyl thiosemicarbazone).

17. The method of claim 16 wherein the second therapeutic includes one or more of Evista (Raloxifene Hydrochloride), Raloxifene Hydrochloride, Tamoxifen Citrate, Abemaciclib, Abraxane (Paclitaxel Albumin-stabilized Nanoparticle Formulation), Ado-Trastuzumab Emtansine, Afinitor (Everolimus), Anastrozole, Aredia (Pamidronate Disodium), Arimidex (Anastrozole), Aromasin (Exemestane), Capecitabine, Cyclophosphamide, Docetaxel, Doxorubicin Hydrochloride, Ellence (Epirubicin Hydrochloride), Epirubicin Hydrochloride, Eribulin Mesylate, Everolimus, Exemestane, 5-FU (Fluorouracil Injection), Fareston (Toremifene), Faslodex (Fulvestrant), Femara (Letrozole), Fluorouracil Injection, Fulvestrant, Gemcitabine Hydrochloride, Gemzar (Gemcitabine Hydrochloride), Goserelin Acetate, Halaven (Eribulin Mesylate), Herceptin (Trastuzumab), Ibrance (Palbociclib), Ixabepilone, Ixempra (Ixabepilone), Kadcyla (Ado-Trastuzumab Emtansine), Kisqali (Ribociclib), Lapatinib Ditosylate, Letrozole, Lynparza (Olaparib), Megestrol Acetate, Methotrexate, Neratinib Maleate, Nerlynx (Neratinib Maleate), Olaparib, Paclitaxel, Paclitaxel Albumin-stabilized Nanoparticle Formulation, Palbociclib, Pamidronate Disodium, Perjeta (Pertuzumab), Pertuzumab, Ribociclib, Tamoxifen Citrate, Taxol (Paclitaxel), Taxotere (Docetaxel), Thiotepa, Toremifene, Trastuzumab, Trexall (Methotrexate), Tykerb (Lapatinib Ditosylate), Verzenio (Abemaciclib), Vinblastine Sulfate, Xeloda (Capecitabine), Zoladex (Goserelin Acetate).

18. The method of claim 1 where the first MEK inhibitor is 3p-Hydroxy-5a, 25R- spirostan-12-(3'-methylphenyl thiosemicarbazone).

19. A pharmaceutical composition for treating a mitogen activated protein kinase kinase/extracellular signal-regulated kinase ("MEK")-induced or an MEK-related pathology in a mammal, comprising:

an effective amount of a first therapeutic; and

an effective amount of a second therapeutic;

wherein the first therapeutic includes a hecogenin-12-thiosemicarbazone, or a pharmaceutically acceptable salt, solvate, ester, amide, clathrate, stereoisomer, enantiomer, prodrug or analog thereof.

20. The pharmaceutical composition of claim 19 wherein the first therapeutic is 3β- Hydroxy-5a, 25R-spirostan-12-(3'-methylphenyl thiosemicarbazone).

Description:
METHODS AND CHEMICALS FOR TREATMENT OF MEK RELATED PATHOLOGIES

[0002] CROSS REFERENCE TO RELATED APPLICATIONS/PRIORITY

[0003] The present invention claims priority to United States Provisional

Patent Application No. 62/557,724 filed September 12, 2017, which is incorporated by reference into the present disclosure as if fully restated herein. Any conflict between the incorporated material and the specific teachings of this disclosure shall be resolved in favor of the latter. Likewise, any conflict between an art-understood definition of a word or phrase and a definition of the word or phrase as specifically taught in this disclosure shall be resolved in favor of the latter.

[0004] BACKGROUND OF INVENTION

[0005] Rat sarcoma small GTPase/rapidly accelerated fibrosarcoma kinase/mitogen activated protein kinase kinase/extracellular signal- regulated kinase (RAS/RAF/MEK/ERK) is an organized conserved eukaryotic signaling pathway. The signaling module is the driver behind paramount cellular processes such as gene transcription, cellular proliferation, and survival. The Ser/Thr kinases MEK1/2 specifically phosphorylate and activate ERK1/2. The activated ERK is dimerized thereafter, regulating various targets in the cytosol and translocating to the nucleus where it phosphorylates a variety of transcription factors regulating gene expression. Despite considerable progress on new molecularly targeted therapies, discovery of more potent, targeted MEK lead inhibitors is still an important therapeutic need, which could save many human lives every year.

[0006] SUMMARY OF THE INVENTION

[0007] Wherefore, it is the object of the present invention to overcome the shortcoming associated with the prior art.

[0008] The sapogenin hecogenin (compound number 1 ) was screened for its anti-breast cancer inhibitory capacity using in vitro assays, including proliferation, cytotoxicity, migration, invasion assays, and western blotting.

l The results identified Cmpd. No. 1 as a potential hit with modest anticancer activities attributed to the concurrent down regulation of mitogen activated protein kinase kinase /extracellular signal-regulated kinase (MEK) distinctive downstream effectors. Guided by in silico 3D- structural insights of the MAPK kinase domain, an extension-strategy was adopted at Cmpd. No. 1 's C-3 and C-12 sites, aimed at the design of novel hecogenin-based analogs with improved target binding affinity. Thirty- three analogs were prepared and tested, among which hecogenin 12-(3'- methylphenyl thiosemicarbazone) (also named 3p-Hydroxy-5a, 25R- spirostan-12-(3'-methylphenyl thiosemicarbazone)) (Analog No. 30) displayed the most potent selective anticancer effects. Analog No. 30 demonstrated antiproliferative, antimigratory and anti-invasive activities at low μΜ level, compared to a negligible effect on the non-tumorigenic MCF- 10A mammary epithelial cells. Reductions in the rate of growth of breast tumor xenografts in athymic nude mice were observed after treatments with Analog No. 30, compared to its parent hecogenin at the same dose regimen, confirming the hit-to-lead promotion of this analog. Hecogenin- 12-thiosemicarbazones, represented by Analog No. 30, is a novel MEK inhibitory lead class for the control breast neoplasms.

[0009] The present invention generally relates to the design and synthesis of hecogenin-based analogs that exhibit anticancer properties and, more specifically, inhibit mitogen activated protein kinase kinase/extracellular signal-regulated kinase (MEK).

[0010] The present invention further relates to hecogenin thiosemicarbazones

(Analog Nos. 15-34) as novel MEK inhibitors and the rational design of semisynthetic hecogenin analogs with improved in vitro anti-breast cancer activities through targeting MEK kinase domain. These compounds, thus, may be used to treat cancer, particularly of the breast, and other MEK- related diseases. The hecogenin thiosemicarbazones disclosed herein are Analog No. 15 (3p-Hydroxy-5a,25R-spirostan-12-(phenyl thiosemicarbazone)), Analog No. 16 (3p-Hydroxy-5a,25R-spirostan-12-(3'- chloroyphenyl thiosemicarbazone)), Analog No. 17 (3p-Hydroxy-5a,25R- spirostan-12-(5'-chlorophenyl thiosemicarbazone)), Analog No. 18 (3β- Hydroxy-5a,25R-spirostan-12-(3',5'-dichlorophenyl thiosemicarbazone)), Analog No. 19 (3p-Hydroxy-5a,25R-spirostan-12-(3\7'-dichlorphenyl thiosemicarbazone)), Analog No. 20 (3p-Hydroxy-5a,25R-spirostan-12-(5'- fluorophenyl thiosemicarbazone)), Analog No. 21 (3p-Hydroxy-5a,25R- spirostan-12-(3'-nitrophenyl thiosemicarbazone)), Analog No. 22 (3β- Hydroxy-5a,25R-spirostan-12-(5'-nitrophenyl thiosemicarbazone)), Analog No. 23 (3p-Hydroxy-5a,25R-spirostan-12-(3'-trifluoromethylphenyl thiosemicarbazone)), Analog No. 24 (3p-Hydroxy-5a,25R-spirostan-12-(5'- trifluoromethylphenyl thiosemicarbazone)), Analog No. 25 (3p-Hydroxy- 5a,25R-spirostan-12-(4',6'-di-triflouromethylphenylthiosemic arbazone)), Analog No. 26 (3p-Hydroxy-5a,25R-spirostan-12-(3'-methoxyphenyl thiosemicarbazone)), Analog No. 27 (3p-Hydroxy-5a,25R-spirostan-12-(5'- methoxyphenyl thiosemicarbazone)), Analog No. 28 (3p-Hydroxy-5a,25R- spirostan-12-(5'-phenoxyphenyl thiosemicarbazone)), Analog No. 29 (3β- Hydroxy-5a,25R-spirostan-12-(3'-ethylphenyl thiosemicarbazone)), Analog No. 30 (3p-Hydroxy-5a,25R-spirostan-12-(3'-methylphenyl thiosemicarbazone)), Analog No. 31 (3p-Hydroxy-5a,25R-spirostan-12-(5'- methylphenyl thiosemicarbazone)), Analog No. 32 (3p-Hydroxy-5a,25R- spirostan-12-(3',5'-dimethylphenyl thiosemicarbazone)), Analog No. 33 (3p-Hydroxy-5a,25R-spirostan-12-(3',7'-dimethylphenyl

thiosemicarbazone)), and Analog No. 34 (3p-Hydroxy-5a,25R-spirostan- 12-(4',5'-methylenedioxyphenyl thiosemicarbazone)).

[001 1 ] The present invention relates to pharmaceutical compositions of a therapeutic (e.g., hecogenin-12-thiosemicarbazones), or a pharmaceutically acceptable salt, solvate, ester, amide, clathrate, stereoisomer, enantiomer, prodrug or analogs thereof, and use of these compositions for the treatment of a MEK-induced pathology, including MEK induced or MEK-related cancer pathologies and MEK induced or MEK-related non-cancer pathologies.

[0012] In some embodiments, the therapeutic, or a pharmaceutically acceptable salt, solvate, or prodrug thereof, is administered as a pharmaceutical composition that further includes a pharmaceutically acceptable excipient.

[0013] In some embodiments, administration of the pharmaceutical composition to a human results in a peak plasma concentration of the therapeutic between 0.05 μΜ-10 μΜ (e.g., between 0.05 μΜ-5 μΜ).

In some embodiments, the peak plasma concentration of the therapeutic is maintained for up to 14 hours. In other embodiments, the peak plasma concentration of the therapeutic is maintained for up to 1 hour.

In some embodiments, the condition is a MEK-induced pathology or MEK-related pathology. In some embodiments, the MEK induced or MEK- related pathology may be a cancer pathology, such as skin cancer, breast cancer, prostate cancer, brain cancer, lung cancer, colorectal cancer, pancreatic cancer, ovarian cancer, liver cancer, bladder cancer, head and neck cancer, esophageal cancer, gastric cancer, or renal cancer. In other embodiments, the MEK-induced or MEK-related pathology may be a non- cancer pathology, such as Alzheimer's disease, osteoarthritis, rheumatoid arthritis, hypertrophic cardiomyopathy, heart failure, stroke, septic shock, organ transplant rejection, and any of the RAS/RAF/MEK/ERK driven pathologies, including Neurofibromatosis Type 1 , Cardiofaciocutaneous syndrome, Noonan syndrome, Noonan syndrome with multiple lentigines (LEOPARD syndrome), Costello syndrome, or Legius syndrome.

In certain embodiments, the MEK induced or MEK-related pathology is mild to moderate MEK induced or MEK-related pathology.

In further embodiments, the MEK induced or MEK-related pathology is moderate to severe MEK induced or MEK-related pathology.

In other embodiments, the therapeutic is administered at a dose that is between 0.05 mg-5 mg/kg weight of the human.

In certain embodiments, the pharmaceutical composition is formulated for oral administration.

In other embodiments, the pharmaceutical composition is formulated for extended release.

In still other embodiments, the pharmaceutical composition is formulated for immediate release.

In some embodiments, the pharmaceutical composition is administered concurrently with one or more additional therapeutic agents for the treatment or prevention of the MEK induced or MEK-related cancer pathology. In some embodiments, the therapeutic, or a pharmaceutically acceptable salt, solvate, or prodrug thereof, is administered as a pharmaceutical composition that further includes a pharmaceutically acceptable excipient.

In some embodiments, administration of the pharmaceutical composition to a human results in a peak plasma concentration of the therapeutic between 0.05 μΜ-10 μΜ (e.g., between 0.05 μΜ-5 μΜ).

In some embodiments, the peak plasma concentration of the therapeutic is maintained for up to 14 hours. In other embodiments, the peak plasma concentration of the therapeutic is maintained for up to 1 hour.

In other embodiments, the therapeutic is administered at a dose that is between 0.05 mg-5 mg/kg weight of the human.

In certain embodiments, the pharmaceutical composition is formulated for oral administration.

In other embodiments, the pharmaceutical composition is formulated for extended release.

In still other embodiments, the pharmaceutical composition is formulated for immediate release.

As used herein, the term "delayed release" includes a pharmaceutical preparation, e.g., an orally administered formulation, which passes through the stomach substantially intact and dissolves in the small and/or large intestine (e.g., the colon). In some embodiments, delayed release of the active agent (e.g., a therapeutic as described herein) results from the use of an enteric coating of an oral medication (e.g., an oral dosage form).

The term an "effective amount" of an agent, as used herein, is that amount sufficient to effect beneficial or desired results, such as clinical results, and, as such, an "effective amount" depends upon the context in which it is being applied.

The terms "extended release" or "sustained release" interchangeably include a drug formulation that provides for gradual release of a drug over an extended period of time, e.g., 6-12 hours or more, compared to an immediate release formulation of the same drug. Preferably, although not necessarily, results in substantially constant blood levels of a drug over an extended time period that are within therapeutic levels and fall within a peak plasma concentration range that is between, for example, 0.05-10 μΜ, 0.1 -10 μΜ, 0.1 -5.0 μΜ, or 0.1 -1 μΜ.

[0033] As used herein, the terms "formulated for enteric release" and "enteric formulation" include pharmaceutical compositions, e.g., oral dosage forms, for oral administration able to provide protection from dissolution in the high acid (low pH) environment of the stomach. Enteric formulations can be obtained by, for example, incorporating into the pharmaceutical composition a polymer resistant to dissolution in gastric juices. In some embodiments, the polymers have an optimum pH for dissolution in the range of approx. 5.0 to 7.0 ("pH sensitive polymers"). Exemplary polymers include methacrylate acid copolymers that are known by the trade name Eudragit ® (e.g., Eudragit ® L100, Eudragit ® S100, Eudragit ® L-30D, Eudragit ® FS 30D, and Eudragit ® L100-55), cellulose acetate phthalate, cellulose acetate trimellitiate, polyvinyl acetate phthalate (e.g., Coateric ® ), hydroxyethylcellulose phthalate, hydroxypropyl methylcellulose phthalate, or shellac, or an aqueous dispersion thereof. Aqueous dispersions of these polymers include dispersions of cellulose acetate phthalate (Aquateric ® ) or shellac (e.g., MarCoat 125 and 125N). An enteric formulation reduces the percentage of the administered dose released into the stomach by at least 50%, 60%, 70%, 80%, 90%, 95%, or even 98% in comparison to an immediate release formulation. Where such a polymer coats a tablet or capsule, this coat is also referred to as an "enteric coating."

[0034] The term "immediate release" includes where the agent (e.g., therapeutic), as formulated in a unit dosage form, has a dissolution release profile under in vitro conditions in which at least 55%, 65%, 75%, 85%, or 95% of the agent is released within the first two hours of administration to, e.g., a human. Desirably, the agent formulated in a unit dosage has a dissolution release profile under in vitro conditions in which at least 50%, 65%, 75%, 85%, 90%, or 95% of the agent is released within the first 30 minutes, 45 minutes, or 60 minutes of administration.

[0035] The term "pharmaceutical composition," as used herein, includes a composition containing a compound described herein (e.g., hecogenin-12- thiosemicarbazones, or any pharmaceutically acceptable salt, solvate, or prodrug thereof), formulated with a pharmaceutically acceptable excipient, and typically manufactured or sold with the approval of a governmental regulatory agency as part of a therapeutic regimen for the treatment of disease in a mammal.

[0036] Pharmaceutical compositions can be formulated, for example, for oral administration in unit dosage form (e.g., a tablet, capsule, caplet, gelcap, or syrup); for topical administration (e.g., as a cream, gel, lotion, or ointment); for intravenous administration (e.g., as a sterile solution free of particulate emboli and in a solvent system suitable for intravenous use); or in any other formulation described herein.

[0037] A "pharmaceutically acceptable excipient," as used herein, includes any ingredient other than the compounds described herein (for example, a vehicle capable of suspending or dissolving the active compound) and having the properties of being nontoxic and non-inflammatory in a patient. Excipients may include, for example: antiadherents, antioxidants, binders, coatings, compression aids, disintegrants, dyes (colors), emollients, emulsifiers, fillers (diluents), film formers or coatings, flavors, fragrances, glidants (flow enhancers), lubricants, preservatives, printing inks, sorbents, suspensing or dispersing agents, sweeteners, or waters of hydration. Exemplary excipients include, but are not limited to: butylated hydroxytoluene (BHT), calcium carbonate, calcium phosphate (dibasic), calcium stearate, croscarmellose, cross-linked polyvinyl pyrrolidone, citric acid, crospovidone, cysteine, ethylcellulose, gelatin, hydroxypropyl cellulose, hydroxypropyl methylcellulose, lactose, magnesium stearate, maltitol, maltose, mannitol, methionine, methylcellulose, methyl paraben, microcrystalline cellulose, polyethylene glycol, polyvinyl pyrrolidone, povidone, pregelatinized starch, propyl paraben, retinyl palmitate, shellac, silicon dioxide, sodium carboxymethyl cellulose, sodium citrate, sodium starch glycolate, sorbitol, starch (corn), stearic acid, stearic acid, sucrose, talc, titanium dioxide, vitamin A, vitamin E, vitamin C, and xylitol.

[0038] The term "pharmaceutically acceptable prodrugs" as used herein, includes those prodrugs of the compounds of the present invention which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and animals with undue toxicity, irritation, allergic response, and the like, commensurate with a reasonable benefit/risk ratio, and effective for their intended use, as well as the zwitterionic forms, where possible, of the compounds of the invention.

[0039] The term "pharmaceutically acceptable salt," as use herein, includes those salts which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and animals without undue toxicity, irritation, allergic response and the like and are commensurate with a reasonable benefit/risk ratio. Pharmaceutically acceptable salts are well known in the art. The salts can be prepared in situ during the final isolation and purification of the compounds of the invention or separately by reacting the free base group with a suitable organic or inorganic acid. Representative acid addition salts include acetate, adipate, alginate, ascorbate, aspartate, benzenesulfonate, benzoate, bisulfate, borate, butyrate, camphorate, camphorsulfonate, citrate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, fumarate, glucoheptonate, glycerophosphate, hemisulfate, heptonate, hexanoate, hydrobromide, hydrochloride, hydroiodide, 2-hydroxy-ethanesulfonate, lactobionate, lactate, laurate, lauryl sulfate, malate, maleate, malonate, methanesulfonate, 2- naphthalenesulfonate, nicotinate, oleate, oxalate, palmitate, pamoate, pectinate, persulfate, 3-phenylpropionate, phosphate, picrate, pivalate, propionate, stearate, succinate, sulfate, tartrate, thiocyanate, toluenesulfonate, undecanoate, valerate salts, and the like. Representative alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium, and the like, as well as nontoxic ammonium, quaternary ammonium, and amine cations, including, but not limited to ammonium, tetramethylammonium, tetraethylammonium, methylamine, dimethylamine, trimethylamine, triethylamine, ethylamine, and the like.

[0040] The terms "pharmaceutically acceptable solvate" or "solvate," as used herein, includes a compound of the invention wherein molecules of a suitable solvent are incorporated in the crystal lattice. A suitable solvent is physiologically tolerable at the administered dose. For example, solvates may be prepared by crystallization, recrystallization, or precipitation from a solution that includes organic solvents, water, or a mixture thereof. Examples of suitable solvents are ethanol, water (for example, mono-, di- and tri-hydrates), /V-methylpyrrolidinone (NMP), dimethyl sulfoxide (DMSO), Λ/,Λ/'-dimethylformamide (DMF), Λ/,Λ/'-dimethylacetamide (DMAC), 1 ,3-dimethyl-2-imidazolidinone (DMEU), 1 ,3-dimethyl-3,4,5,6- tetrahydro-2-(1 H)-pyrimidinone (DMPU), acetonitrile (ACN), propylene glycol, ethyl acetate, benzyl alcohol, 2-pyrrolidone, benzyl benzoate, and the like. When water is the solvent, the solvate is referred to as a "hydrate."

[0041 ] The term "prevent," as used herein, includes prophylactic treatment or treatment that prevents one or more symptoms or conditions of a disease, disorder, or conditions described herein (e.g., a MEK induced or MEK- related cancer pathology). Treatment can be initiated, for example, prior to ("pre-exposure prophylaxis") or following ("post-exposure prophylaxis") an event that precedes the onset of the disease, disorder, or conditions. Treatment that includes administration of a compound of the invention, or a pharmaceutical composition thereof, can be acute, short-term, or chronic. The doses administered may be varied during the course of preventive treatment.

[0042] The term "prodrug," as used herein, includes compounds which are rapidly transformed in vivo to the parent compound of the above formula. Prodrugs also encompass bioequivalent compounds that, when administered to a human, lead to the in vivo formation of therapeutic. Preferably, prodrugs of the compounds of the present invention are pharmaceutically acceptable.

[0043] As used herein, and as well understood in the art, "treatment" includes an approach for obtaining beneficial or desired results, such as clinical results. Beneficial or desired results can include, but are not limited to, alleviation or amelioration of one or more symptoms or conditions; diminishment of extent of disease, disorder, or condition; stabilized (i.e. not worsening) state of disease, disorder, or condition; preventing spread of disease, disorder, or condition; delay or slowing the progress of the disease, disorder, or condition; amelioration or palliation of the disease, disorder, or condition; and remission (whether partial or total), whether detectable or undetectable. "Treatment" can also mean prolonging survival as compared to expected survival if not receiving treatment. As used herein, the terms "treating" and "treatment" can also include delaying the onset of, impeding or reversing the progress of, or alleviating either the disease or condition to which the term applies, or one or more symptoms of such disease or condition.

[0044] The term "unit dosage forms" includes physically discrete units suitable as unitary dosages for human subjects and other mammals, each unit containing a predetermined quantity of active material calculated to produce the desired therapeutic effect, in association with any suitable pharmaceutical excipient or excipients.

[0045] As used herein, the term "plasma concentration" includes the amount of therapeutic present in the plasma of a treated subject (e.g., as measured in a rabbit using an assay described below or in a human).

[0046] The invention is related to compositions and methods of treating a mitogen activated protein kinase kinase/extracellular signal-regulated kinase ("MEK")-induced or an MEK-related pathology in a mammal, comprising administering a pharmaceutical composition containing an effective amount of a first therapeutic, wherein the first therapeutic includes a first MEK inhibitor, or a pharmaceutically acceptable salt, solvate, ester, amide, clathrate, stereoisomer, enantiomer, prodrug or analog thereof. According to a further embodiment, the mammal is human According to a further embodiment, the first MEK inhibitor is (3p,5a,25R)- 3-Hydroxyspirostan-12-one. According to a further embodiment, the first MEK inhibitor is a Hecogenin-12-thiosemicarbazones. According to a further embodiment, the first MEK inhibitor is one of 3p-Hydroxy-5a,25R- spirostan-12-(phenyl thiosemicarbazone), 3p-Hydroxy-5a,25R-spirostan- 12-(3'-chloroyphenyl thiosemicarbazone), 3p-Hydroxy-5a,25R-spirostan- 12-(5'-chlorophenyl thiosemicarbazone), 3p-Hydroxy-5a,25R-spirostan-12- (3',5'-dichlorophenyl thiosemicarbazone), 3p-Hydroxy-5a,25R-spirostan- 12-(3',7'-dichlorphenyl thiosemicarbazone), 3p-Hydroxy-5a,25R-spirostan- 12-(5'-fluorophenyl thiosemicarbazone), 3p-Hydroxy-5a,25R-spirostan-12- (3'-nitrophenyl thiosemicarbazone), 3p-Hydroxy-5a,25R-spirostan-12-(5'- nitrophenyl thiosemicarbazone), 3p-Hydroxy-5a,25R-spirostan-12-(3'- trifluoromethylphenyl thiosemicarbazone), 3p-Hydroxy-5a,25R-spirostan- 12-(5'-trifluoromethylphenyl thiosemicarbazone), 3p-Hydroxy-5a,25R- spirostan-12-(4',6'-di-triflouromethylphenylthiosemicarbazon e), 3β- Hydroxy-5a,25R-spirostan-12-(3'-methoxyphenyl thiosemicarbazone), 3β- Hydroxy-5a,25R-spirostan-12-(5'-methoxyphenyl thiosemicarbazone), 3β- Hydroxy-5a,25R-spirostan-12-(5'-phenoxyphenyl thiosemicarbazone), 3β- Hydroxy-5a,25R-spirostan-12-(3'-ethylphenyl thiosemicarbazone), 3β- Hydroxy-5a,25R-spirostan-12-(3'-methylphenyl thiosemicarbazone), 3β- Hydroxy-5a,25R-spirostan-12-(5'-methylphenyl thiosemicarbazone), 3β- Hydroxy-5a,25R-spirostan-12-(3',5'-dimethylphenyl thiosemicarbazone), 3β-Hydroxy-5α,25R-spirostan-12-(3',7'-dimethylphenyl

thiosemicarbazone), and 3β-Hydroxy-5α,25R-spirostan-12-(4',5'- methylenedioxyphenyl thiosemicarbazone). According to a further embodiment, the pharmaceutical composition contains a second therapeutic for the MEK-induced pathology or MEK-related pathology distinct from the first therapeutic. According to a further embodiment, the second therapeutic is not a MEK inhibitor. According to a further embodiment, the second therapeutic is one of second therapeutic could be one of Evista (Raloxifene Hydrochloride), Raloxifene Hydrochloride, Tamoxifen Citrate, Abemaciclib, Abraxane (Paclitaxel Albumin-stabilized Nanoparticle Formulation), Ado-Trastuzumab Emtansine, Afinitor (Everolimus), Anastrozole, Aredia (Pamidronate Disodium), Arimidex (Anastrozole), Aromasin (Exemestane), Capecitabine, Cyclophosphamide, Docetaxel, Doxorubicin Hydrochloride, Ellence (Epirubicin Hydrochloride), Epirubicin Hydrochloride, Eribulin Mesylate, Everolimus, Exemestane, 5- FU (Fluorouracil Injection), Fareston (Toremifene), Faslodex (Fulvestrant), Femara (Letrozole), Fluorouracil Injection, Fulvestrant, Gemcitabine Hydrochloride, Gemzar (Gemcitabine Hydrochloride), Goserelin Acetate, Halaven (Eribulin Mesylate), Herceptin (Trastuzumab), Ibrance (Palbociclib), Ixabepilone, Ixempra (Ixabepilone), Kadcyla (Ado- Trastuzumab Emtansine), Kisqali (Ribociclib), Lapatinib Ditosylate, Letrozole, Lynparza (Olaparib), Megestrol Acetate, Methotrexate, Neratinib Maleate, Nerlynx (Neratinib Maleate), Olaparib, Paclitaxel, Paclitaxel Albumin-stabilized Nanoparticle Formulation, Palbociclib, Pamidronate Disodium, Perjeta (Pertuzumab), Pertuzumab, Ribociclib, Tamoxifen Citrate, Taxol (Paclitaxel), Taxotere (Docetaxel), Thiotepa, Toremifene, Trastuzumab, Trexall (Methotrexate), Tykerb (Lapatinib Ditosylate), Verzenio (Abemaciclib), Vinblastine Sulfate, Xeloda (Capecitabine), Zoladex (Goserelin Acetate). According to a further embodiment, the second therapeutic includes a second MEK inhibitor. According to a further embodiment, the MEK-induced or MEK-related pathology is cancer or pre cancer state. According to a further embodiment, the cancer is one of a skin cancer, breast cancer, prostate cancer, brain cancer, lung cancer, colorectal cancer, pancreatic cancer, ovarian cancer, liver cancer, bladder cancer, head and neck cancer, esophageal cancer, gastric cancer, and renal cancer. According to a further embodiment, the MEK-induced or MEK-related pathology is a non-cancer pathology. According to a further embodiment, the non-cancer pathology is one of Alzheimer's disease, osteoarthritis, rheumatoid arthritis, hypertrophic cardiomyopathy, heart failure, stroke, septic shock, organ transplant rejection, and an RAS/RAF/MEK/ERK driven pathology. According to a further embodiment, the RAS/RAF/MEK/ERK driven pathology includes one of Neurofibromatosis Type 1 , Cardiofaciocutaneous syndrome, Noonan syndrome, Noonan syndrome with multiple lentigines (LEOPARD syndrome), Costello syndrome, and Legius syndrome. According to a further embodiment, the mammal is human, the MEK induced or MEK related pathology is a cancer, the pharmaceutical composition contains a second therapeutic for the cancer, and the first MEK inhibitor is a Hecogenin-12-thiosemicarbazones. According to a further embodiment, the cancer is a breast cancer. According to a further embodiment, the first MEK inhibitor is 3p-Hydroxy-5a, 25R-spirostan-12-(3'-methylphenyl thiosemicarbazone). According to a further embodiment, the second therapeutic includes one or more of Evista (Raloxifene Hydrochloride), Raloxifene Hydrochloride, Tamoxifen Citrate, Abemaciclib, Abraxane (Paclitaxel Albumin-stabilized Nanoparticle Formulation), Ado- Trastuzumab Emtansine, Afinitor (Everolimus), Anastrozole, Aredia (Pamidronate Disodium), Arimidex (Anastrozole), Aromasin (Exemestane), Capecitabine, Cyclophosphamide, Docetaxel, Doxorubicin Hydrochloride, Ellence (Epirubicin Hydrochloride), Epirubicin Hydrochloride, Eribulin Mesylate, Everolimus, Exemestane, 5-FU (Fluorouracil Injection), Fareston (Toremifene), Faslodex (Fulvestrant), Femara (Letrozole), Fluorouracil Injection, Fulvestrant, Gemcitabine Hydrochloride, Gemzar (Gemcitabine Hydrochloride), Goserelin Acetate, Halaven (Eribulin Mesylate), Herceptin (Trastuzumab), Ibrance (Palbociclib), Ixabepilone, Ixempra (Ixabepilone), Kadcyla (Ado-Trastuzumab Emtansine), Kisqali (Ribociclib), Lapatinib Ditosylate, Letrozole, Lynparza (Olaparib), Megestrol Acetate, Methotrexate, Neratinib Maleate, Nerlynx (Neratinib Maleate), Olaparib, Paclitaxel, Paclitaxel Albumin-stabilized Nanoparticle Formulation, Palbociclib, Pamidronate Disodium, Perjeta (Pertuzumab), Pertuzumab, Ribociclib, Tamoxifen Citrate, Taxol (Paclitaxel), Taxotere (Docetaxel), Thiotepa, Toremifene, Trastuzumab, Trexall (Methotrexate), Tykerb (Lapatinib Ditosylate), Verzenio (Abemaciclib), Vinblastine Sulfate, Xeloda (Capecitabine), Zoladex (Goserelin Acetate). According to a further embodiment, the first MEK inhibitor is 3p-Hydroxy-5a, 25R-spirostan-12- (3'-methylphenyl thiosemicarbazone).

[0047] The invention further relates to methods and pharmaceutical compositions for treating a mitogen activated protein kinase kinase/extracellular signal-regulated kinase ("MEK")-induced or an MEK- related pathology in a mammal, comprising an effective amount of a first therapeutic and an effective amount of a second therapeutic, wherein the first therapeutic includes a hecogenin-12-thiosemicarbazone, or a pharmaceutically acceptable salt, solvate, ester, amide, clathrate, stereoisomer, enantiomer, prodrug or analog thereof. According to a further embodiment, the first therapeutic is 3p-Hydroxy-5a, 25R-spirostan- 12-(3'-methylphenyl thiosemicarbazone).

[0048] DESCRIPTION OF THE DRAWINGS

[0049] The accompanying drawings, which are incorporated in and constitute a part of the specification, illustrate various embodiments of the invention, and together with the general description of the invention described above and the detailed description of the drawings described below, serve to illustrate the key features of the invention. Colors described in the brief description refer to colors in the figures filed in United States Provisional Patent Application No. 62/557,724 filed September 12, 2017. The invention will now be described, using the examples, with reference to the accompanying drawings in which:

FIG. 1 shows the effect of Hecogenin (Cmpd. No. 1 ) treatments on the proliferation of multiple human breast cancer cell lines. Displayed is the mean percent cell viability at different concentrations of Cmpd. No. 1 after 72 h incubation. The horizontal dashed line shows the calculated IC50 ±SEM for each cell line.

FIGS. 2A-2C show the overview of the binding of Cmpd. No. 1 in green (ball and stick cartoon or CPK format) with MEK kinase domain (PDB: 3EQF) represented as a ribbon diagram. The protein is oriented as for the canonical kinase domain, with the N-terminal lobe at the top and the C-terminal lobe at the bottom. The three figures are from a similar vantage point, displayed in space-filling model the good fitting of Cmpd. No. 1 within the identified grid. Cmpd. No. 1 Displayed favorable H-bond interactions with residues Met146 and Lys192, while enough space around C-3 hydroxyl and C-12 ketone likely to accommodate subsequent structure extensions. FIG. 2D is a structural model of Cmpd. No. 1 .

FIG. 3 shows a general semisynthetic route to hecogenin esters (Analog Nos. 2-9), referred to herein as Scheme 1 . Reagents and conditions: (i) acid chloride, pyridine, DMAP, rt, overnight.

FIG. 4 shows semisynthesis of hecogenin phenylsemicarbazones (Analog Nos. 10-14) and hecogenin thiosemicarbazones (Analog Nos. 15-34), referred to herein as Scheme 2. Reagents and conditions: (i) Semicarbazide.HCI, EtOH, Pyridine, rt, 6 h. (ii) Semicarbazide/thiosemicarbazide, EtOH, rt, overnight.

FIG. 5 shows effects of analog number 30 treatments on multiple human breast cancer cell lines and the human non-tumorigenic mammary epithelial cells MCF-10A. Displayed is the mean percent cell viability at different concentrations of Analog No. 30 after treatment incubation period. The horizontal dashed line shows calculated IC50 ±SEM of Analog No. 30 for each cell line. [0055] FIGS. 6A-6C show chemical models of Analog Nos. 2 - 34.

[0056] FIGS 7A - 7B together show in vitro antiproliferative activity

(IC50 (μΜ) ±SE) of Compound Number 1 and Analog Nos. 2-34 against a panel of human breast cancer cell lines, referred to herein as Table 1 .

[0057] FIGS 8A-8D show binding mode of Analog No. 30 at the MEK kinase domain (PDB: 3EQF). FIG. 8A: Chemical structure of Analog No. 30. FIG. 8B: Ribbon diagram of the binding pose of Analog No. 30 (maroon ball and stick cartoon) at the MEK kinase domain. This spacefilling model revealed the good fitting of Analog No. 30 within the proximity of Phe209 and Cys207 residues satisfying the C-12 extension tactic. FIG. 8C: Binding pose of Analog No. 30 within the MEK kinase domain using CPK format. FIG. 8D: Space filling model showing the protein target canonical kinase domain, with the N-terminal lobe at the top and the C- terminal lobe at the bottom. All parts are from a similar vantage point.

[0058] FIGS 9A-9C show (FIG. 9A) Overview of MEK domain (PDB:

3EQF) represented as a ribbon diagram in complex with Cmpd. No. 1 in green and Analog No. 30 in maroon with ball and stick cartoon format. (FIG. 9B) Superposition of Cmpd. No. 1 in green and Analog No. 30 in maroon with ball and stick cartoon format. (FIG. 9C) Superposition of K252A (MEK ATP-competitive inhibitor in orange) with Analog No. 30 in maroon.

[0059] FIG. 10 shows percent cytotoxicity of Cmpd. No. 1 and Analog

No. 30 evaluated by LDH release from MDA-MB-231 cells, referred to herein as Table 2.

[0060] FIGS. 11A-11 D show effects of Cmpd. No. 1 and Analog No. 30 on the TNBC MDA-MB-231 cells migration capacity using WHA. (FIGS. 11A and 11 B) Representative microscopic images of created wounds at zero time and 24 h post-incubation with vehicle as negative control or various treatment concentrations. (FIGS. 11 C and 11 D) Dose response curve of Cmpd. No. 1 and Analog No. 30 treatments versus percent wound closure.

[0061 ] FIGS 12A-12C show effects of Analog No. 30, compared to its parent Cmpd. No. 1 against the TNBC MDA-MB-231 cells invasive capacity using CultreCoat® cell invasion assay. (FIG. 12A) Representative microscopic images of upper chambers (non-invasive cell density) with the vehicle, Cmpd. No. 1 or Analog No. 30. (FIGS. 12B and 12C) Dose response effect of Cmpd. No. 1 and Analog No. 30 treatments versus average percent cell invasion.

FIGS. 13A-13B show (FIG. 13A) overview of the MAPK pathway. (FIG. 13B) Western blot analyses of in vitro vehicle control and Analog No. 30 treatments in the TNBC MDA-MB-231 cells. Data show downregulation of activated MAPK kinase and downstream effectors, ERK and MSK in both treatment groups compared to vehicle control.

FIGS 14A-14C show In vivo anticancer activities of Cmpd. No. 1 and Analog No. 30 against MDA-MB-231 /GFP breast cancer cells in athymic nude mouse xenograft model. (FIG. 14A): Effects of Cmpd. No. 1 and Analog No. 30 treatments on the mean tumor volume throughout the study compared to vehicle control group. (FIG. 14B) Percent tumor growth (TG) in treatment groups, compared to vehicle control group calculated at the study end. (FIG. 14C) Pictorial representation of mice from control and treatment groups at the study end.

FIG. 15 shows in vivo anticancer activity of Cmpd. No. 1 and Analog No. 30 against the TNBC MDA-MB-231 cells in nude mouse xenograft model, referred to herein as Table 3.

[0065] DETAILED DESCRIPTION OF INVENTION

[0066] The present invention will be understood by reference to the following detailed description, which should be read in conjunction with the appended drawings. It is to be appreciated that the following detailed description of various embodiments is by way of example only and is not meant to limit, in any way, the scope of the present invention. In the summary above, in the following detailed description, in the claims below, and in the accompanying drawings, reference is made to particular features (including method steps) of the present invention. It is to be understood that the disclosure of the invention in this specification includes all possible combinations of such particular features, not just those explicitly described. For example, where a particular feature is disclosed in the context of a particular aspect or embodiment of the invention or a particular claim, that feature can also be used, to the extent possible, in combination with and/or in the context of other particular aspects and embodiments of the invention, and in the invention generally. The term "comprises" and grammatical equivalents thereof are used herein to mean that other components, ingredients, steps, etc. are optionally present. For example, an article "comprising" (or "which comprises") components A, B, and C can consist of (i.e., contain only) components A, B, and C, or can contain not only components A, B, and C but also one or more other components. Where reference is made herein to a method comprising two or more defined steps, the defined steps can be carried out in any order or simultaneously (except where the context excludes that possibility), and the method can include one or more other steps which are carried out before any of the defined steps, between two of the defined steps, or after all the defined steps (except where the context excludes that possibility).

The term "at least" followed by a number is used herein to denote the start of a range beginning with that number (which may be a range having an upper limit or no upper limit, depending on the variable being defined). For example "at least 1 " means 1 or more than 1 . The term "at most" followed by a number is used herein to denote the end of a range ending with that number (which may be a range having 1 or 0 as its lower limit, or a range having no lower limit, depending upon the variable being defined). For example, "at most 4" means 4 or less than 4, and "at most 40%" means 40% or less than 40%. When, in this specification, a range is given as "(a first number) to (a second number)" or "(a first number)-(a second number)," this means a range whose lower limit is the first number and whose upper limit is the second number. For example, 25 to 100 mm means a range whose lower limit is 25 mm, and whose upper limit is 100 mm. The embodiments set forth the below represent the necessary information to enable those skilled in the art to practice the invention and illustrate the best mode of practicing the invention. In addition, the invention does not require that all the advantageous features and all the advantages need to be incorporated into every embodiment of the invention. [0068] Turning now to FIGS. 1 -15, a brief description concerning the various components of the present invention will now be briefly discussed. Hecogenin (Cmpd. No. 1 ), a ubiquitous spirostanic steroidal sapogenin in sisal wastes, has been previously described for its anticancer activity. Cmpd. No. 1 was screened in vitro for ability to inhibit breast cancer proliferation, migration, and invasion. The antiproliferative effect of Cmpd. No. 1 was assessed against a panel of six human mammary carcinoma cell lines displaying diverse molecular and phenotypic characteristics. For instance, estrogen receptor (ER a ) is expressed in the luminal A-type MCF- 7 and T-47D cell lines; while SKBR-3 and the luminal B-type BT-474 express human epidermal growth factor receptor 2 (HER2). In addition, the claudin low MDA-MB-231 and the basal MDA-MB-468 were negative for the triple receptors; ER a , HER2 and progesterone receptor (PR) and so alternatively named, triple negative breast cancer (TNBC) cell lines. Hecogenin (Cmpd. No. 1 ) exhibited modest antiproliferative activity with IC 50 values in the mid μΜ level (FIG. 1 ). Thus, the inventors hypothesized that Cmpd. No. 1 targets a common pathway(s) involved in the proliferation and survival of breast cancer cells.

[0069] Hecogenin-3-O-acetate's is known to the inventors to modulate the extracellular signal-regulated kinases (ERK1/2) phosphorylation. ERK is the only downstream substrate for the non-receptor hydrophilic protein mitogen-activated protein kinase/extracellular signal-regulated kinase (MEK). MEK1/2 phosphorylates, and activates ERK1/2-mediated oncogenic proliferation and differentiation pathways. The inventors conducted a computational docking experiment using Glide 5.8 module in standard mode to explore the virtual binding modes of Cmpd. No. 1 and hence explore potential future rational design of more potent semisynthetic analogs. Careful inspection of the prepared dual-specific MEK protein kinase revealed its resemblance to alternative protein kinases that possess a multifunctional, small N-terminal lobe, protein kinase cleft and large C-terminal lobe comprising several conserved a-helices and β- strands. Hecogenin (Cmpd. No. 1 ) was anchored within the generated grid constraints of MEK cavity with a notable docking score of -7.9. The space-filling model assumed Cmpd. No. 1 was in an extended spatial conformation, consistent with the general horizontal dimensions of the pocket, leaving the C-3 hydroxyl and C-12 ketone enough spaces to accommodate structure extensions (FIG. 2). Interestingly, Cmpd. No. 1 displayed a favorable H-bond bridge by the C-3 OH group with the gatekeeper Met146 carbonyl backbone allocated in the hinge region, while the rear pyran oxygen displayed a H-bond interaction with Lys192 terminal amino in the catalytic loop. The inventors then hypothesized that Cmpd. No. 1 was to be a potential MEK inhibitory scaffold amenable for semisynthetic optimizations at C-3 hydroxyl and C-12 ketone owing to the vacant room prevailed behind.

The first optimization approach for Cmpd. No. 1 targeted the C-3 spacer domain via esterification of C-3 secondary alcohol. This was implemented through base-catalyzed esterification reaction of Cmpd. No. 1 with various aromatic acid chlorides (FIG. 3, Scheme 1 ), generating the corresponding ester Analog Nos. 2-9.

Purified products were fully characterized by 1 D and 2D NMR spectroscopy and mass spectrometry. For instance, 3-O-hecogenin benzoyl ester (Analog No. 2) displayed a downfield shift of H-3 to δ 4.90 (+1 .3 ppm), along with its corresponding carbon C-3 by +4.0 ppm (5 75.0), compared to the parent Cmpd. No. 1 , confirming the C-3 esterification.

The human triple negative breast cancer (TNBC) MDA-MB-231 cell-line expresses mutant RAS and RAF proteins, and predictably showed high levels of phosphorylated ERK (p-ERK) expression, indicating active MAPK pathway. Additionally, MDA-MB-231 cells were sensitive to treatment with Cmpd. No. 1 as demonstrated using the MTT proliferation assay (FIG. 1 ). Thus, semisynthesized esters were evaluated for their antiproliferative potency on the panel of six breast adenocarcinoma cell lines, while their SAR was configured according to their activity against MDA-MB-231 cells. Results demonstrated that ester analogs showed no improvement in Cmpd. No. 1 's antiproliferative activity (Table 1 , FIG 7A). This may be attributed, at least in-part, to potential hydrophobic interactions with the proximity amino acids that replaced the free C-3 OH interactions, which may stipulate the significance of the C-3 OH group to anchorage at the target kinase's hinge region. Subsequent optimizations maintained a free C-3 OH group and targeted the C-12 spacer in Cmpd. No. 1 to probe additional binding interactions. An extension strategy was adopted using both a hydrophilic linker capable to interplay H-bond bridges with the target, and a lipophilic functionalized substituent directed towards the target hydrophobic cleft. Hence, a cross-coupling reaction using various phenyl semicarbazides and phenyl thiosemicarbazides appropriately functionalized on the aromatic ring was successfully achieved (FIG. 4, Scheme 2).

The aimed products were purified, isolated in moderate to excellent yields and predicted structures elucidated using 1 D and 2D NMR spectroscopy and mass spectrometry. Careful inspection of the 13 C- PENDANT NMR spectrum of hecogenin-12-(phenyl semicarbazone) (Analog No. 10) and hecogenin-12-(phenyl thiosemicarbazone) (Analog No. 15) revealed the replacement of Cmpd. No. 1 's C-12 ketone signal (5 C 213.4) with new ones at 5 C 158.5 or 159.9, corresponding to their new C- 12 imine carbonyls, respectively. The new imine carbonyl carbons showed 3 J HMBC cross interactions with the C-18 methyl proton singlets (δ Η 0.98- 1 .00). Moreover, the integrity of urea or thiourea fragment was based on their C- carbonyl or thiocarbonyl at 5 C 154.6 or 5 C 176.3, respectively. Meanwhile, the broad singlet signals at δ 8.50-9.50 were ascribable to the two aza-methines of the urea fragment, manifesting 2 J HMBC correlations with the C- carbonyl and thiocarbonyl or the C-2 ' quaternary aromatic carbons. In addition, one of the aza-methines showed a 3 J HMBC correlation with the C-12 imine carbonyl. While E and Z-geometrical isomers are possible for the non-symmetric semi-and thio-semicarbazone products, it is interesting to note that evidence for such isomeric mixtures was not observed by 1 D NMR experiments, suggesting the presence of only one isomer. The 1 H- 1 H NOESY spectrum indicated the exclusive oxime double bond E-geometry orientation in Analog No. 30, based on the cross peak interactions between the hydrazine NH singlet (δ 8.70) and H 2 - 1 1 methylene proton at δ 1 .92. This assignment was in agreement with the literature, which the preference to the less sterically hindered E-isomer product of such reactions. Thus, the same double bond geometry was also assumed in rest of analogs.

Analog Nos. 10-34 were evaluated for their antiproliferative activities against the TNBC MDA-MB-231 cell line. The improved activity of hecogenin-12-phenylsemicarbazone (Analog No. 10), compared to the parent Cmpd. No. 1 (IC50 = 9.2μΜ) encouraged additional optimizations. Various analogs with a substituted phenyl ring that featured different steric, hydrophobic, and electronic properties were synthesized and tested to measure inhibitor properties. For instance, the electron withdrawing hybrids represented by Analog Nos. 1 1 -13; m-chlorophenyl, p- chlorophenyl and m-trifluoromethyl phenyl semicarbazones, respectively, were semisynthesized and tested. The chlorinated analogs were more potent than Analog No. 10 (FIGS. 7A-7B, Table 1 ), even though the positional effect of the CI atom seems to have a significant role in the activity. The para-positioned chlorinated analog appeared more potent than its mefa-conger (FIGS. 7A-7B, Table 1 ). Unexpectedly, the meta- substituted Analog No. 13, bearing a trifluoromethyl phenyl moiety displayed a significant activity decrease (IC50 = 14.0 μΜ). Interestingly, while examining the urea linker optimal span, the tosyl semicarbazone Analog No. 14 revealed significantly reduced activity compared to Analog No. 10 (FIGS. 7A-7B, Table 1 ), suggesting the four-atom hydrazine carbothioamide hydrophilic linker is optimal for target affinity and subsequent cellular activity. This conclusion could also be supported by the literature use of "soft" donor atoms, as sulfur in thiosemicarbazone scaffolds for good anticancer activity. Hecogenin-12-phenyl thiosemicarbazone (Analog No. 15) exhibited the expected improved potency (IC 50 = 7.8 μΜ), compared to its phenyl semicarbazone counterpart Analog No. 10, supporting the activity preference for thiosemicarbazones. The impact of electronic effects of different chloro, fluoro, nitro, and trifluoromethyl phenyl-substituted thiosemicarbazones Analog Nos. 16-25 were then studied. The 3 ' -trifluoromethyl-bearing Analog No. 23 exhibited the most attained potency of this group (IC 50 = 2.1 μΜ). In addition, the electron-donating functionalities were also explored through synthesizing and biological testing of methoxy (Analog Nos. 26- 27), phenoxy (Analog No. 28), ethyl (Analog No. 29), methyl (Analog Nos. 30-33) and methylenedioxy (Analog No. 34) phenyl thiosemicarbazone analogs. Notably, the 3 ' -methyl Analog No. 30 exhibited the greatest potency against a panel of breast cancer cell lines (FIGS. 7A-7B, Table 1 , and FIG 5), highlighting the requirement of an optimal mono-substituted, hydrophobic, sterically tolerable phenyl substituent. Hence, Analog No. 30 was selected for a comprehensive activity study.

[0076] Molecular docking of Analog No. 30 within the prepared MEK kinase domain showed that it is quite closely overlaps with its parent Cmpd. No. 1 , though with a better extension towards the hydrophobic pocket, affording an excellent docking score of -9.0. The imine nitrogen displayed a H-bond bridge with the amino acid Ser194, meanwhile the E- configuration enabled the substituted phenyl thio-semicarbazone fragment to fit well within a hydrophobic pocket clustered with Val127, Ile141 , Cys207, Leu74 and Met143 residues, generating an excellent hydrophobic interaction. Interestingly, the distal phenyl moiety in Analog No. 30 displayed parallel-displaced stacking with Phe209 allocated within the Asp-Phe-Gly (DFG) motif, while the o/f/?o-methyl functionality predominantly contributed a hydrophobic interaction with Cys207- methylene. The thiourea linker formed a H-bond interaction with the Lys97's positively charged terminal amino group (FIGS. 8A-8D).

[0077] To add further justification to the docking results, the native ligand of the MEK crystal structure (PDB: 3EQF) was docked into its ATP binding site, applying the same parameters which have been used for docking Analog No. 30 (FIGS. 9A-9C). The bound conformation of co-crystallized ligand was obtained with a good root mean square displacement (RMSD) of 0.2 A, suggesting the robustness of the docking experiments. In the investigated MEK crystal structure PDB 3EQF, 30 demonstrated partial overlay with almost the same interactions (FIGS. 9A-9C). Together, the in siiico data suggested the potential of Analog No. 30 as a suitable MEK inhibitory hit appropriate for further validation.

[0078] Many therapies, while killing the bulk of cancer cells, may ultimately fail to discriminate normal healthy cells from malignant cells. Accordingly, the anticancer selectivity of Analog No. 30 was evaluated using the human mammary epithelial cell line MCF-10A. These cells are immortalized and non-tumorigenic with some features of normal breast epithelium, including lack of anchorage-independent growth and dependence on growth factors and hormones for proliferation and survival. Cells were exposed to various concentrations of Analog No. 30 and viability was assessed. The results showed that treatment of cells with Analog No. 30 over a range of concentrations (1 -20 μΜ) had no significant effect on MCF-10A viability, compared to their respective vehicle control-treated group, reinforcing the Analog No. 30's selective anticancer effect towards malignant breast cells.

[0079] Measurement of intracellular content leakage through impaired plasma membrane has been widely used to assess chemical cytotoxicity. Lactate dehydrogenase (LDH) is a soluble cytoplasmic enzyme expressed in almost all cells, and is released into extracellular space in respond to plasma membrane destruction. Therefore, the detection of LDH in the culture medium can be used as a marker for cytotoxicity. Thus, the Cayman's LDH cytotoxicity assay kit was used to evaluate the ability of Analog No. 30 to induce LDH release in MDA-MB-231 cells in comparison to its parent Cmpd. No. 1 . Four different doses of tested compounds were used to assess targeted cell death. Hecogenin (Cmpd. No. 1 ) showed 24.6% cytotoxicity at the maximum tested dose (100 μΜ), which is more than three-fold its reported antiproliferative IC50. Analog No. 30 demonstrated 5.9% cytotoxicity at the maximum tested dose (10 μΜ), a concentration which is more than five-fold its reported antiproliferative IC 50 (FIG. 10, Table 2).

[0080] Although cancer cell proliferation is often regarded as the most important aspect of cancer progression, other important cellular processes play a crucial role in cancer progression and metastasis, including migration and invasion. Hence, interfering with both processes could have positive impacts on patient survival. A wound healing (WHA) and CultreCoat invasion assay were used to determine the inhibitory effects of Cmpd. No. 1 on the migration and invasion capacities of the highly metastatic MDA-MB-231 cells. The WHA assay clearly demonstrated that after 24 hours, Cmpd. No. 1 significantly repressed migration of MDA-MB- 231 cells towards the denuded zone in a dose response fashion (FIGS.1 1 A-1 1 C) with a calculated IC 50 of 2.5μΜ.

[0081 ] The effects of Cmpd. No. 1 and Analog No. 30 on MDA-MB-231 cell invasion across the extracellular matrix was also tested using four concentrations of drugs. Analog No. 30 significantly inhibited cell invasion, with maximum percent invasion inhibition calculated as 92.4% at 5 μΜ (maximum tested dose) (FIGS. 12A-12C). In comparison, treatment with Cmpd. No. 1 at the maximum tested dose (80 μΜ) significantly inhibited cell invasion, with 86.2% invasion inhibition. The calculated IC50 for Analog No. 30 was 2.2 μΜ, compared to 37.1 μΜ for the parent Cmpd. No. 1 .

[0082] MAPK regulates diverse cellular programs that coordinately regulate cell proliferation, differentiation, motility, and survival. The wide range of functions regulated by the MAPK is mediated through phosphorylation of several downstream substrates, including members of a family of protein kinases termed MAPK-activated protein kinases (MAPKAPKs) as mitogen- and stress-activated kinases (MSKs) and MAPK-interacting kinases (MNKs).

[0083] Accordingly, to confirm the MEK inhibitory activity of Analog No. 30,

Western blot analysis was implemented using MDA-MB-231 cell lysate, following administration of two different doses of Cmpd. No. 1 and Analog No. 30, along with a vehicle control treatment. The phosphorylation levels of the specific MEK downstream effector, ERK (MAPK) was significantly reduced in both Cmpd. No. 1 and Analog No. 30 treatment groups, compared to vehicle control treatment (FIGS. 13A-13B). Collectively, western blot analysis results strongly support the ability of Analog No. 30 to inhibit MEK catalytic activity, thus interfering with MAPK cellular functions.

[0084] Significant in vitro activity of Analog No. 30 against MDA-MB-231 cells

(FIGS.7A-7B, Table 1 ) motivated a subsequent in vivo study to assess its anticancer efficacy compared with its parent Cmpd. No. 1 in a pertinent breast cancer xenograft model. Athymic nude Foxn1 nu /Foxn1 + mice orthotopically- transplanted with MDA-MB-231 /GFP cells were used. Five days after tumor cell implantation, solid tumors became palpable. Mice then were intraperitoneally administrated equivalent 10 mg/kg/3X week doses of Cmpd. No. 1 (group I) or Analog No. 30 (group II), while the control group received vehicle only. Treatments were continued for twenty- eight days. Mice were observed and body weights were monitored per dosing as a preliminary toxicity assessment. Both treatments demonstrated no overt signs of either toxicity or significant body weight change, compared to the vehicle-control group. Consistent with the in vitro lack of toxicity against the non-tumorigenic MCF10A mammary epithelial cells, nude mice showed good tolerance to both treatments. Tumor growth was monitored and quantified. On the basis of caliper measurements, the treatment groups demonstrated a significant reduction in tumor growth by the end of the study, compared to the vehicle control group. Hecogenin (Cmpd. No. 1 ) treatment reduced tumor growth by 57% while treatment with analog Analog No. 30 resulted in a 78% reduction in tumor growth (FIG. 15, Table 3). Ultimately, Analog No. 30 significantly suppressed cell proliferation and attenuated tumor growth in this orthotopic model of TNBC and is therefore considered a lead compound.

[0085] Pharmaceutical Compositions: The methods described herein can also include the administrations of pharmaceutically acceptable compositions that include the therapeutic, or a pharmaceutically acceptable salt, solvate, or prodrug thereof. When employed as pharmaceuticals, any of the present compounds can be administered in the form of pharmaceutical compositions. These compositions can be prepared in a manner well known in the pharmaceutical art, and can be administered by a variety of routes, depending upon whether local or systemic treatment is desired and upon the area to be treated. Administration may be topical, parenteral, intravenous, intra-arterial, subcutaneous, intramuscular, intracranial, intraorbital, ophthalmic, intraventricular, intracapsular, intraspinal, intracisternal, intraperitoneal, intranasal, aerosol, by suppositories, or oral administration.

[0086] This invention also includes pharmaceutical compositions which can contain one or more pharmaceutically acceptable carriers. In making the pharmaceutical compositions of the invention, the active ingredient is typically mixed with an excipient, diluted by an excipient or enclosed within such a carrier in the form of, for example, a capsule, sachet, paper, or other container. When the excipient serves as a diluent, it can be a solid, semisolid, or liquid material (e.g., normal saline), which acts as a vehicle, carrier or medium for the active ingredient. Thus, the compositions can be in the form of tablets, powders, lozenges, sachets, cachets, elixirs, suspensions, emulsions, solutions, syrups, and soft and hard gelatin capsules. As is known in the art, the type of diluent can vary depending upon the intended route of administration. The resulting compositions can include additional agents, such as preservatives.

[0087] The therapeutic agents of the invention can be administered alone, or in a mixture, in the presence of a pharmaceutically acceptable excipient or carrier. The excipient or carrier is selected on the basis of the mode and route of administration. Suitable pharmaceutical carriers, as well as pharmaceutical necessities for use in pharmaceutical formulations will be used. In preparing a formulation, the active compound can be milled to provide the appropriate particle size prior to combining with the other ingredients. If the active compound is substantially insoluble, it can be milled to a particle size of less than 200 mesh. If the active compound is substantially water soluble, the particle size can be adjusted by milling to provide a substantially uniform distribution in the formulation, e.g. about 40 mesh.

[0088] Examples of suitable excipients are lactose, dextrose, sucrose, sorbitol, mannitol, starches, gum acacia, calcium phosphate, alginates, tragacanth, gelatin, calcium silicate, microcrystalline cellulose, polyvinylpyrrolidone, cellulose, water, syrup, and methyl cellulose. The formulations can additionally include: lubricating agents such as talc, magnesium stearate, and mineral oil; wetting agents; emulsifying and suspending agents; preserving agents such as methyl- and propylhydroxy- benzoates; sweetening agents; and flavoring agents. Other exemplary excipients are described in Handbook of Pharmaceutical Excipients, 8 th Edition, Sheskey et al., Eds., Pharmaceutical Press (2017), which is incorporated by reference.

[0089] The methods described herein can include the administration of a therapeutic, or prodrugs or pharmaceutical compositions thereof, or other therapeutic agents. Exemplary therapeutics include those that concurrently down regulate mitogen activated protein kinase kinase /extracellular signal-regulated kinase (including hecogenin-12- thiosemicarbazones).

[0090] The pharmaceutical compositions can be formulated so as to provide immediate, extended, or delayed release of the active ingredient after administration to the patient by employing procedures known in the art.

[0091 ] The compositions can be formulated in a unit dosage form, each dosage containing, e.g., 0.1 -500 mg of the active ingredient. For example, the dosages can contain from about 0.1 mg to about 50 mg, from about 0.1 mg to about 40 mg, from about 0.1 mg to about 20 mg, from about 0.1 mg to about 10 mg, from about 0.2 mg to about 20 mg, from about 0.3 mg to about 15 mg, from about 0.4 mg to about 10 mg, from about 0.5 mg to about 1 mg; from about 0.5 mg to about 100 mg, from about 0.5 mg to about 50 mg, from about 0.5 mg to about 30 mg,, from about 0.5 mg to about 20 mg, from about 0.5 mg to about 10 mg, from about 0.5 mg to about 5 mg; from about 1 mg from to about 50 mg, from about 1 mg to about 30 mg,, from about 1 mg to about 20 mg, from about 1 mg to about 10 mg, from about 1 mg to about 5 mg; from about 5 mg to about 50 mg, from about 5 mg to about 20 mg, from about 5 mg to about 10 mg; from about 10 mg to about 100 mg, from about 20 mg to about 200 mg, from about 30 mg to about 150 mg, from about 40 mg to about 100 mg, from about 50 mg to about 100 mg of the active ingredient, from about 50 mg to about 300 mg, from about 50 mg to about 250 mg, from about 100 mg to about 300 mg, or , from about 100 mg to about 250 mg of the active ingredient. For preparing solid compositions such as tablets, the principal active ingredient is mixed with one or more pharmaceutical excipients to form a solid bulk formulation composition containing a homogeneous mixture of a compound of the present invention. When referring to these bulk formulation compositions as homogeneous, the active ingredient is typically dispersed evenly throughout the composition so that the composition can be readily subdivided into equally effective unit dosage forms such as tablets and capsules. This solid bulk formulation is then subdivided into unit dosage forms of the type described above containing from, for example, 0.1 to about 500 mg of the active ingredient of the present invention.

[0092] Compositions for Oral Administration: The pharmaceutical compositions contemplated by the invention include those formulated for oral administration ("oral dosage forms"). Oral dosage forms can be, for example, in the form of tablets, capsules, a liquid solution or suspension, a powder, or liquid or solid crystals, which contain the active ingredient(s) in a mixture with non-toxic pharmaceutically acceptable excipients. These excipients may be, for example, inert diluents or fillers (e.g., sucrose, sorbitol, sugar, mannitol, microcrystalline cellulose, starches including potato starch, calcium carbonate, sodium chloride, lactose, calcium phosphate, calcium sulfate, or sodium phosphate); granulating and disintegrating agents (e.g., cellulose derivatives including microcrystalline cellulose, starches including potato starch, croscarmellose sodium, alginates, or alginic acid); binding agents (e.g., sucrose, glucose, sorbitol, acacia, alginic acid, sodium alginate, gelatin, starch, pregelatinized starch, microcrystalline cellulose, magnesium aluminum silicate, carboxymethylcellulose sodium, methylcellulose, hydroxypropyl methylcellulose, ethylcellulose, polyvinylpyrrolidone, or polyethylene glycol); and lubricating agents, glidants, and antiadhesives (e.g., magnesium stearate, zinc stearate, stearic acid, silicas, hydrogenated vegetable oils, or talc). Other pharmaceutically acceptable excipients can be colorants, flavoring agents, plasticizers, humectants, buffering agents, and the like.

[0093] Formulations for oral administration may also be presented as chewable tablets, as hard gelatin capsules wherein the active ingredient is mixed with an inert solid diluent (e.g., potato starch, lactose, microcrystalline cellulose, calcium carbonate, calcium phosphate or kaolin), or as soft gelatin capsules wherein the active ingredient is mixed with water or an oil medium, for example, peanut oil, liquid paraffin, or olive oil. Powders, granulates, and pellets may be prepared using the ingredients mentioned above under tablets and capsules in a conventional manner using, e.g., a mixer, a fluid bed apparatus or a spray drying equipment.

[0094] Controlled release compositions for oral use may be constructed to release the active drug by controlling the dissolution and/or the diffusion of the active drug substance. Any of a number of strategies can be pursued in order to obtain controlled release and the targeted plasma concentration vs time profile. In one example, controlled release is obtained by appropriate selection of various formulation parameters and ingredients, including, e.g., various types of controlled release compositions and coatings. Thus, the drug is formulated with appropriate excipients into a pharmaceutical composition that, upon administration, releases the drug in a controlled manner. Examples include single or multiple unit tablet or capsule compositions, oil solutions, suspensions, emulsions, microcapsules, microspheres, nanoparticles, patches, and liposomes. In certain embodiments, compositions include biodegradable, pH, and/or temperature-sensitive polymer coatings.

[0095] Dissolution or diffusion controlled release can be achieved by appropriate coating of a tablet, capsule, pellet, or granulate formulation of compounds, or by incorporating the compound into an appropriate matrix. A controlled release coating may include one or more of the coating substances mentioned above and/or, e.g., shellac, beeswax, glycowax, castor wax, carnauba wax, stearyl alcohol, glyceryl monostearate, glyceryl distearate, glycerol palmitostearate, ethylcellulose, acrylic resins, dl- polylactic acid, cellulose acetate butyrate, polyvinyl chloride, polyvinyl acetate, vinyl pyrrolidone, polyethylene, polymethacrylate, methylmethacrylate, 2-hydroxymethacrylate, methacrylate hydrogels, 1 ,3 butylene glycol, ethylene glycol methacrylate, and/or polyethylene glycols. In a controlled release matrix formulation, the matrix material may also include, e.g., hydrated methylcellulose, carnauba wax and stearyl alcohol, carbopol 934, silicone, glyceryl tristearate, methyl acrylate-methyl methacrylate, polyvinyl chloride, polyethylene, and/or halogenated fluorocarbon.

[0096] The liquid forms in which the compounds and compositions of the present invention can be incorporated for administration orally include aqueous solutions, suitably flavored syrups, aqueous or oil suspensions, and flavored emulsions with edible oils such as cottonseed oil, sesame oil, coconut oil, or peanut oil, as well as elixirs and similar pharmaceutical vehicles.

[0097] Compositions suitable for oral mucosal administration (e.g., buccal or sublingual administration) include tablets, lozenges, and pastilles, where the active ingredient is formulated with a carrier, such as sugar, acacia, tragacanth, or gelatin and glycerine.

[0098] Coatings: The pharmaceutical compositions formulated for oral delivery, such as tablets or capsules of the present invention can be coated or otherwise compounded to provide a dosage form affording the advantage of delayed or extended release. The coating may be adapted to release the active drug substance in a predetermined pattern (e.g., in order to achieve a controlled release formulation) or it may be adapted not to release the active drug substance until after passage of the stomach, e.g., by use of an enteric coating (e.g., polymers that are pH-sensitive ("pH controlled release"), polymers with a slow or pH-dependent rate of swelling, dissolution or erosion ("time-controlled release"), polymers that are degraded by enzymes ("enzyme-controlled release" or "biodegradable release") and polymers that form firm layers that are destroyed by an increase in pressure ("pressure-controlled release")). Exemplary enteric coatings that can be used in the pharmaceutical compositions described herein include sugar coatings, film coatings (e.g., based on hydroxypropyl methylcellulose, methylcellulose, methyl hydroxyethylcellulose, hydroxypropylcellulose, carboxymethylcellulose, acrylate copolymers, polyethylene glycols and/or polyvinylpyrrolidone), or coatings based on methacrylic acid copolymer, cellulose acetate phthalate, hydroxypropyl methylcellulose phthalate, hydroxypropyl methylcellulose acetate succinate, polyvinyl acetate phthalate, shellac, and/or ethylcellulose. Furthermore, a time delay material such as, for example, glyceryl monostearate or glyceryl distearate, may be employed.

[0099] For example, the tablet or capsule can comprise an inner dosage and an outer dosage component, the latter being in the form of an envelope over the former. The two components can be separated by an enteric layer which serves to resist disintegration in the stomach and permit the inner component to pass intact into the duodenum or to be delayed in release. [0100] When an enteric coating is used, desirably, a substantial amount of the drug is released in the lower gastrointestinal tract.

[0101 ] In addition to coatings that effect delayed or extended release, the solid tablet compositions may include a coating adapted to protect the composition from unwanted chemical changes (e.g., chemical degradation prior to the release of the active drug substance). The coating may be applied on the solid dosage form in a similar manner as that described in Encyclopedia of Pharmaceutical Technology, vols. 5 and 6, Eds. Swarbrick and Boyland, 2000.

[0102] Parenteral Administration: Within the scope of the present invention are also parenteral depot systems from biodegradable polymers. These systems are injected or implanted into the muscle or subcutaneous tissue and release the incorporated drug over extended periods of time, ranging from several days to several months. Both the characteristics of the polymer and the structure of the device can control the release kinetics which can be either continuous or pulsatile. Polymer-based parenteral depot systems can be classified as implants or microparticles. The former are cylindrical devices injected into the subcutaneous tissue whereas the latter are defined as spherical particles in the range of 10 - 100 pm. Extrusion, compression or injection molding are used to manufacture implants whereas for microparticles, the phase separation method, the spray-drying technique and the water-in-oil-in-water emulsion techniques are frequently employed. The most commonly used biodegradable polymers to form microparticles are polyesters from lactic and/or glycolic acid, e.g. poly(glycolic acid) and poly(L-lactic acid) (PLG/PLA microspheres). Of particular interest are in situ forming depot systems, such as thermoplastic pastes and gelling systems formed by solidification, by cooling, or due to the sol-gel transition, cross-linking systems and organogels formed by amphiphilic lipids. Examples of thermosensitive polymers used in the aforementioned systems include, N- isopropylacrylamide, poloxamers (ethylene oxide and propylene oxide block copolymers, such as poloxamer 188 and 407), poly(N-vinyl caprolactam), poly(siloethylene glycol), polyphosphazenes derivatives and PLGA-PEG-PLGA. Mucosal Drug Delivery: Mucosal drug delivery (e.g., drug delivery via the mucosal linings of the nasal, rectal, vaginal, ocular, or oral cavities) can also be used in the methods described herein. Methods for oral mucosal drug delivery include sublingual administration (via mucosal membranes lining the floor of the mouth), buccal administration (via mucosal membranes lining the cheeks), and local delivery.

Oral transmucosal absorption is generally rapid because of the rich vascular supply to the mucosa and allows for a rapid rise in blood concentrations of the therapeutic.

For buccal administration, the compositions may take the form of, e.g., tablets, lozenges, etc. formulated in a conventional manner. Permeation enhancers can also be used in buccal drug delivery. Exemplary enhancers include 23-lauryl ether, aprotinin, azone, benzalkonium chloride, cetylpyridinium chloride, cetyltrimethylammonium bromide, cyclodextrin, dextran sulfate, lauric acid, lysophosphatidylcholine, methol, methoxysalicylate, methyloleate, oleic acid, phosphatidylcholine, polyoxyethylene, polysorbate 80, sodium EDTA, sodium glycholate, sodium glycodeoxycholate, sodium lauryl sulfate, sodium salicylate, sodium taurocholate, sodium taurodeoxycholate, sulfoxides, and alkyl glycosides. Bioadhesive polymers have extensively been employed in buccal drug delivery systems and include cyanoacrylate, polyacrylic acid, hydroxypropyl methylcellulose, and poly methacrylate polymers, as well as hyaluronic acid and chitosan.

Liquid drug formulations (e.g., suitable for use with nebulizers and liquid spray devices and electrohydrodynamic (EHD) aerosol devices) can also be used. Other methods of formulating liquid drug solutions or suspension suitable for use in aerosol devices are known to those of skill in the art.

Formulations for sublingual administration can also be used, including powders and aerosol formulations. Exemplary formulations include rapidly disintegrating tablets and liquid-filled soft gelatin capsules.

Dosing Regimes: The present methods for treating MEK-induced or MEK-related pathologies are carried out by administering a therapeutic for a time and in an amount sufficient to result in decreased tumor growth or decreased tumor size, for example, in embodiments of MEK-induced or MEK-related cancer pathologies.

The amount and frequency of administration of the compositions can vary depending on, for example, what is being administered, the state of the patient, and the manner of administration. In therapeutic applications, compositions can be administered to a patient suffering from MEK-induced or MEK-related pathology in an amount sufficient to relieve or least partially relieve the symptoms of the MEK-induced or MEK-related pathology and its complications. The dosage is likely to depend on such variables as the type and extent of progression of the MEK-induced or MEK-related pathology, the severity of the MEK-induced or MEK-related pathology, the age, weight and general condition of the particular patient, the relative biological efficacy of the composition selected, formulation of the excipient, the route of administration, and the judgment of the attending clinician. Effective doses can be extrapolated from dose- response curves derived from in vitro or animal model test system. An effective dose is a dose that produces a desirable clinical outcome by, for example, improving a sign or symptom of the MEK-induced or MEK- related pathology or slowing its progression.

The amount of therapeutic per dose can vary. For example, a subject can receive from about 0.1 g/kg to about 10,000 g/kg. Generally, the therapeutic is administered in an amount such that the peak plasma concentration ranges from 150 nM-250 μΜ.

Exemplary dosage amounts can fall between 0.1 -5000 pg/kg, 100- 1500 pg/kg, 100-350 pg/kg, 340-750 pg/kg, or 750-1000 pg/kg. Exemplary dosages can 0.25, 0.5, 0.75, 1°, or 2 mg/kg. In another embodiment, the administered dosage can range from 0.05-5 mmol of therapeutic (e.g., 0.089-3.9 mmol) or 0.1 -50 pmol of therapeutic (e.g., 0.1 - 25 pmol or 0.4-20 pmol).

The plasma concentration of therapeutic can also be measured according to methods known in the art. Exemplary peak plasma concentrations of therapeutic can range from 0.05-10 μΜ, 0.1 -10 μΜ, 0.1 - 5.0 μΜ, or 0.1 -1 μΜ. Alternatively, the average plasma levels of therapeutic can range from 400-1200 μΜ (e.g., between 500-1000 μΜ) or between 50-250 μΜ (e.g., between 40-200 μΜ). In some embodiments where sustained release of the drug is desirable, the peak plasma concentrations (e.g., of therapeutic) may be maintained for 6-14 hours, e.g., for 6-12 or 6-10 hours. In other embodiments where immediate release of the drug is desirable, the peak plasma concentration (e.g., of therapeutic) may be maintained for, e.g., 30 minutes.

The frequency of treatment may also vary. The subject can be treated one or more times per day with therapeutic (e.g., once, twice, three, four or more times) or every so-many hours (e.g., about every 2, 4, 6, 8, 12, or 24 hours). Preferably, the pharmaceutical composition is administered 1 or 2 times per 24 hours. The time course of treatment may be of varying duration, e.g., for two, three, four, five, six, seven, eight, nine, ten or more days. For example, the treatment can be twice a day for three days, twice a day for seven days, twice a day for ten days. Treatment cycles can be repeated at intervals, for example weekly, bimonthly or monthly, which are separated by periods in which no treatment is given. The treatment can be a single treatment or can last as long as the life span of the subject (e.g., many years).

Kits

Any of the pharmaceutical compositions of the invention described herein can be used together with a set of instructions, i.e., to form a kit. The kit may include instructions for use of the pharmaceutical compositions as a therapy as described herein. For example, the instructions may provide dosing and therapeutic regimes for use of the compounds of the invention to reduce symptoms and/or underlying cause of the MEK-induced or MEK-related pathology.

Second Therapeutics. According to a prophetic example, the MEK inhibitor will be paired with an current or established therapeutic for a particular MEK-induced or MEK-related pathology offering anticipated additive or synergistic effects.. For example, with the MEK induced or MEK related pathology of cancer, such as breast cancer, a first therapeutic would be an MEK inhibitor, such as Analog No 30, and a second therapeutic could be one or more of Evista (Raloxifene Hydrochloride), Raloxifene Hydrochloride, Tamoxifen Citrate, Drugs Approved to Treat Breast Cancer, Abemaciclib, Abraxane (Paclitaxel Albumin-stabilized Nanoparticle Formulation), Ado-Trastuzumab Emtansine, Afinitor (Everolimus), Anastrozole, Aredia (Pamidronate Disodium), Arimidex (Anastrozole), Aromasin (Exemestane), Capecitabine, Cyclophosphamide, Docetaxel, Doxorubicin Hydrochloride, Ellence (Epirubicin Hydrochloride), Epirubicin Hydrochloride, Eribulin Mesylate, Everolimus, Exemestane, 5-FU (Fluorouracil Injection), Fareston (Toremifene), Faslodex (Fulvestrant), Femara (Letrozole), Fluorouracil Injection, Fulvestrant, Gemcitabine Hydrochloride, Gemzar (Gemcitabine Hydrochloride), Goserelin Acetate, Halaven (Eribulin Mesylate), Herceptin (Trastuzumab), Ibrance (Palbociclib), Ixabepilone, Ixempra (Ixabepilone), Kadcyla (Ado- Trastuzumab Emtansine), Kisqali (Ribociclib), Lapatinib Ditosylate, Letrozole, Lynparza (Olaparib), Megestrol Acetate, Methotrexate, Neratinib Maleate, Nerlynx (Neratinib Maleate), Olaparib, Paclitaxel, Paclitaxel Albumin-stabilized Nanoparticle Formulation, Palbociclib, Pamidronate Disodium, Perjeta (Pertuzumab), Pertuzumab, Ribociclib, Tamoxifen Citrate, Taxol (Paclitaxel), Taxotere (Docetaxel), Thiotepa, Toremifene, Trastuzumab, Trexall (Methotrexate), Tykerb (Lapatinib Ditosylate), Verzenio (Abemaciclib), Vinblastine Sulfate, Xeloda (Capecitabine), Zoladex (Goserelin Acetate).

The invention illustratively disclosed herein suitably may explicitly be practiced in the absence of any element which is not specifically disclosed herein. While various embodiments of the present invention have been described in detail, it is apparent that various modifications and alterations of those embodiments will occur to and be readily apparent those skilled in the art. However, it is to be expressly understood that such modifications and alterations are within the scope and spirit of the present invention, as set forth in the appended claims. Further, the invention(s) described herein is capable of other embodiments and of being practiced or of being carried out in various other related ways. In addition, it is to be understood that the phraseology and terminology used herein is for the purpose of description and should not be regarded as limiting. The use of "including," "comprising," or "having" and variations thereof herein is meant to encompass the items listed thereafter and equivalents thereof as well as additional items while only the terms "consisting of" and "consisting only of" are to be construed in the limitative sense.