WO2016003886A1 - Compositions de 4-aminoquinoline et méthodes d'utilisation - Google Patents

Compositions de 4-aminoquinoline et méthodes d'utilisation Download PDF

Info

Publication number
WO2016003886A1
WO2016003886A1 PCT/US2015/038287 US2015038287W WO2016003886A1 WO 2016003886 A1 WO2016003886 A1 WO 2016003886A1 US 2015038287 W US2015038287 W US 2015038287W WO 2016003886 A1 WO2016003886 A1 WO 2016003886A1
Authority
WO
WIPO (PCT)
Prior art keywords
cancer
cell
compound
jtc
inhibitor
Prior art date
Application number
PCT/US2015/038287
Other languages
English (en)
Inventor
Neil BEEHARRY
Paul Beckett
Jonathan M. Rothberg
Henri Lichenstein
Marylens HERNANDEZ
Original Assignee
Lam Therapeutics, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Lam Therapeutics, Inc. filed Critical Lam Therapeutics, Inc.
Publication of WO2016003886A1 publication Critical patent/WO2016003886A1/fr

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/47064-Aminoquinolines; 8-Aminoquinolines, e.g. chloroquine, primaquine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/13Amines
    • A61K31/145Amines having sulfur, e.g. thiurams (>N—C(S)—S—C(S)—N< and >N—C(S)—S—S—C(S)—N<), Sulfinylamines (—N=SO), Sulfonylamines (—N=SO2)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/506Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim not condensed and containing further heterocyclic rings

Definitions

  • the present disclosure relates to compositions and methods for the treatment of mTOR diseases, disorders or conditions, and for the treatment of proliferative disorders including cancer.
  • the mammalian target of rapamycin (mTOR) pathway is an important cellular signaling pathway that is involved in multiple physiological functions, including cell growth, cell proliferation, metabolism, protein synthesis, and autophagy.
  • mTOR is a kinase that integrates intracellular and extracellular cues that signal the levels of amino acids, stress, oxygen, energy, and growth factors and regulates the cellular response to these environment cues.
  • mTOR deregulation has been implicated in a wide range of disorders and diseases, including cancer, obesity, diabetes, and neurodegeneration. Certain components of the mTOR pathway have been explored as drug targets for treating some of these diseases. However, therapeutic efficacy has been limited, for example, in the treatment of some cancers, and some mTOR inhibitors have been shown to have an adverse effect on metabolism.
  • Lymphangioleiomyomatosis is a multisystem disease affecting 30-40% of women with tuberous sclerosis complex (TSC), an often-fatal disease which is characterized by the widespread proliferation of abnormal smooth muscle-like cells that grow aberrantly in the lung.
  • TSC tuberous sclerosis complex
  • LAM cells The proliferation of these cells leads to the formation of cysts in the lungs and fluid- filled cystic structures in the axial lymphatics (referred to as lymphangioleiomyomas).
  • LAM cells can form tumors.
  • the abnormal proliferation of LAM cells is caused at least in part by an inactivating mutation in one of the tuberous sclerosis complex tumor suppressor genes, TSC 1 or TSC2.
  • the TSC genes are negative regulators of mTOR.
  • LAM cells show constitutive activation of mTOR and many of the mTOR-related kinases such as Akt, Erk, S6K1 and S6.
  • the present disclosure relates to methods of treating an mTOR-related disease, disorder or condition and methods for treating proliferative diseases, disorders, and conditions using 4-aminoquinoline compounds identified herein as having cytotoxic and/or anti-proliferative activity.
  • the 4-aminoquinoline compound is referred to as JTC-801.
  • the present disclosure provides methods for treating an mTOR disease, disorder or condition in a subject in need thereof, the method including administering to the subject a therapeutically effective amount of a pharmaceutical composition comprising JTC-801, or a pharmaceutically acceptable salt, solvate, clathrate, hydrate, polymorph, metabolite, prodrug, analog or derivative thereof.
  • the pharmaceutical composition includes JTC-801.
  • the mTOR disease, disorder or condition can be selected from genetic tumor syndromes, neurological diseases, fragile X syndrome, Down syndrome, Rett syndrome, epilepsy, autism/neurodevelopmental disorders, neurodegenerative diseases, metabolic diseases or proliferative disorders, including cancer.
  • the proliferative disorder is a cancer that is not known to be mediated by hyper activation of mTOR.
  • the neurodegenerative disease can be Alzheimer's, Parkinson's, Huntington's disease, amyotrophic lateral sclerosis, or frontotemporal dementia.
  • the metabolic disease can be obesity, diabetes such as Type I or Type II diabetes, lipotoxicity, hyperlipidemia, hyperglycemia, insulin resistance, hyperglycemia,
  • hyperinsulinemia hyperinsulinemia, hypoinsulinemic, fatty liver disease, or cirrhosis.
  • the proliferative disorder can be cancer.
  • the cancer has not previously been linked to constitutive activation of mTOR.
  • the cancer can be brain cancer, glioma, sarcoma, breast cancer, lung cancer, non-small-cell lung cancer, mesothelioma, appendiceal cancer, genitourinary cancers, renal cell carcinoma, prostate cancer, bladder cancer, testicular cancer, penile cancer, cervical cancer, ovarian cancer, von Hippel Lindau disease, head and neck cancer, gastrointestinal cancer, hepatocellular carcinoma, gallbladder cancer, esophageal cancer, gastric cancer, colorectal cancer, pancreatic cancer,
  • neuroendocrine tumors thyroid tumor, pituitary tumor, adrenal tumor, hematological malignancy, Hodgkin's lymphoma, Non-Hodgkin's lymphoma, mantle cell lymphoma, myeloma, B-cell lymphoma, leukemia or melanoma.
  • the mTOR disease, disorder or condition can be Tuberous Sclerosis (TSC) and/or Lymphangioleiomyomatosis (LAM).
  • TSC Tuberous Sclerosis
  • LAM Lymphangioleiomyomatosis
  • the disclosure provides methods of treating cancer in a subject in need thereof, the methods comprising administering to the subject JTC-801.
  • the cancer is selected from the group consisting of colon cancer, prostate cancer, lung cancer, breast cancer, pancreatic cancer, a leukemia, and a neuroglioma.
  • an amount of JTC-801 is administered effective alleviate one or more symptoms of the cancer, for example an amount effective to inhibit the proliferation of the cancer cells and/or to slow the growth or reduce the size of a solid tumor, or to prevent the spread or further spread of the cancer to other tissues of the subject.
  • the method can further include administering to the subject one or more additional therapies or therapeutic agents as part of a therapeutic regimen.
  • the one or more additional therapeutic agents can be selected from the group consisting of an mTOR inhibitor, a statin, preferably simvastatin, an anti-estrogen therapy, doxycycline, a tyrosine kinase inhibitor, a src inhibitor, an autophagy inhibitor (e.g.
  • the one or more additional therapeutic agents is a tyrosine kinase inhibitor.
  • the tyrosine kinase inhibitor is selected from imatinib and pazopanib.
  • the one or more additional therapies can be selected from chemotherapy, radiation therapy, hormonal therapy, anti-estrogen therapy, biological response modifier treatment, bone marrow transplantation, gene therapy, and surgery.
  • the one or more additional therapies is selected from anti-estrogen therapy or hormone therapy.
  • the therapeutically effective amount of the pharmaceutical composition can be an amount effective to reduce the size of a tumor, to reduce tumor number, improve the subject's pulmonary function as measured by forced vital capacity (FVC) and forced expiratory volume (FEV1) or an amount effective to reduce the size or amount of pleural effusion detectable by radiologic examination.
  • FVC forced vital capacity
  • FEV1 forced expiratory volume
  • the administered dose of JTC-801 can be about 0.001 mg/kg to about 1000 mg/kg, more preferably about 0.01 mg/kg to about 100 mg/kg, more preferably about 0.1 mg/kg to about 10 mg/kg.
  • the administered dose of imatinib or pazopanib can be 10 to 100 micrograms, more preferably 50 to 250 micrograms, more preferably 100 to 500 micrograms (0.1 to 0.5 milligrams), more preferably 500 to 1000 micrograms (0.5 to 1 milligrams) or more preferably 1000 to 2000 micrograms (1 to 2 milligrams), more preferably less than 2 milligrams.
  • the present disclosure also provides a pharmaceutical composition including a therapeutically effective amount of JTC-801, or a pharmaceutically acceptable salt, solvate, clathrate, hydrate, polymorph, metabolite, prodrug, analog or derivative thereof, the amount being effective to treat an mTOR disease, disorder or condition in a subject in need of such treatment.
  • the disorder is a proliferative disorder.
  • the JTC-801, or a pharmaceutically acceptable salt, solvate, clathrate, hydrate, polymorph, metabolite, prodrug, analog or derivative thereof is JTC-801.
  • the one or more additional therapeutic agents can be selected from the group consisting of an mTOR inhibitor, a statin, preferably simvastatin, an anti-estrogen therapy, doxycycline, a tyrosine kinase inhibitor, a src inhibitor, an autophagy inhibitor (e.g.
  • the one or more additional therapeutic agents are a tyrosine kinase inhibitor.
  • the tyrosine kinase inhibitor is selected from imatinib or pazopanib.
  • Tyrosine Kinase inhibitor imatinib (Gleevec) is currently used in the treatment of multiple cancers, most notably Philadelphia chromosome-positive chronic myelogenous leukemia. Imatinib is dosed orally once a day for doses 400-600 mg or twice a day for a 800 mg dose. Combination therapy with imatinib has been investigated in many clinical trials. For example, everolismus in combination with imatinib was investigated for gastrointestinal stromal tumors. A dacarbazine/imatinib combination was used in the treatment for thyroid cancer and solid tumors. Panobinostat and imatinib was used in treating Leukemia. Docetaxel and imatinib combinations or letrozole and imatinib combinations were evaluated in breast cancer. Combinations with imatinib and hydroxyurea were examined in glioblastoma and gliosarcoma.
  • pazopanib (Votrient), a potent and selective multi-targeted receptor tyrosine kinase inhibitor is approved for the treatment of renal cell carcinoma and soft tissue carcinoma. Oral administration of 800 mg once a day is recommended.
  • Combination therapy with pazopanib has also been investigated in many clinical trials. For example, combination therapy of pazopanib with doxorubicin or temsirolimus for advanced solid tumors has been investigated. Also, nivolumab in combination with pazopanib has been investigated for renal cell carcinoma. Gemcitabine/ pazopanib combinations have been examined in lung cancer. Lastly, pazopanib and carboplatin were evaluated in advanced solid malignancies.
  • the disclosure provides methods of treating a proliferative disease or disorder by administering JTC-801 in combination with an additional therapeutic agent.
  • the disclosure provides methods of treating of proliferative disorder selected from chronic myelogenous leukemia, gastrointestinal stromal tumors and acute lymphoblastic leukemia, the method comprising administering JTC-801 in combination with imatinib.
  • the disclosure provides methods of treating of proliferative disorder selected from breast cancer and lung cancer, the method comprising administering JTC-801 in combination with fluvestrant.
  • the disclosure provides methods of treating of proliferative disorder selected from breast cancer and pancreatic cancer, the method comprising administering JTC-801 in combination with disulfiram.
  • the disclosure provides methods of treating of proliferative disorder selected from renal cell carcinoma, advanced soft tissue sarcomas, colon, lung, and breast cancers, the method comprising administering JTC-801 in combination with pazopanib.
  • FIG.1 is a series of graphs illustrating HTS concentration response curves in MEF-EV and MEF-TSC2 cells.
  • FIG. 2 is a graph illustrating concentration response curves of JTC-801 in various cancer cell lines.
  • the indicated cancer cell lines cells were treated with JTC-801 (19.5- ⁇ ) for 72h before assayed for viability. IC50 values were calculated and shown.
  • FIG. 3 is a graph illustrating HTS screening of unapproved compounds vs. pazopanib to identify synergistic drugs reveals JTC-801.
  • MEF-EV cells were treated with a library of unapproved compounds in the presence or absence of pazopanib. Each dot shown is the viability value of an individual unapproved compound in the absence (X-axis) or presence (Y axis) of pazopanib. Dark dots indicate significant interactions, of which JTC-801 is shown (arrow).
  • FIG. 4 is a graph illustrating combination treatment of JTC-801 and pazopanib synergistically reduces ELT-3 cell viability.
  • ELT-3 cells were treated with either JTC-801 or pazopanib alone or in combination. Cells were treated for 72h before assayed for viability.
  • FIG. 5 is a graph illustrating combination treatment of JTC-801 and pazopanib synergistically reduces cancer cell line viability.
  • a panel of cancer cell lines is treated with JTC-801 (19.5-10000nM) in the presence or absence of pazopanib (5000nM). Cells were treated for 72h before assayed for viability.
  • a representative graph displaying synergistic activity in HI 299 cells is shown.
  • FIG. 6 is a graph illustrating HTS screening of unapproved compounds vs. disulfiram to identify synergistic drugs reveals JTC-801.
  • MEF-EV cells were treated with a library of unapproved compounds in the presence or absence of disulfiram ( ⁇ ). Each dot shown is the viability value of an individual unapproved compound in the absence (X-axis) or presence (Y axis) of disulfiram. Dark dots indicate significant interactions, of which JTC-801 is shown (arrow).
  • FIG. 7 is a graph illustrating HTS screening of unapproved compounds vs. faslodex to identify synergistic drugs reveals JTC-801.
  • MEF-EV cells were treated with a library of unapproved compounds in the presence or absence of faslodex (5000nM). Each dot shown is the viability value of an individual unapproved compound in the absence (X-axis) or presence (Y axis) of faslodex. Dark dots indicate significant interactions, of which JTC-801 is shown (arrow).
  • FIG. 8 is a graph illustrating combination treatment of JTC-801 and imatinib synergistically reduces ELT-3 cell viability.
  • ELT-3 cells were treated with either JTC-801 or imatinib alone or in combination. Cells were treated for 72h before assayed for viability.
  • FIG. 9A is a graph illustrating the cancer tissue hits corresponding to JTC-801 identified via computational analysis.
  • FIG. 9B depicts bar graphs by calculating the percentage of tissue-specific cell lines with a negative dds score.
  • the X axis of the bar graphs represent the tissue that the analyzed cancer cells with a negative score correspond to drawing showing structural analogs of JTC-801 : JTC-801-1, JTC-801 -2, JTC-801 -3 and JTC-801 -4.
  • FIG. 10 depicts drawing of structural analogs of JTC-801 : JTC-801-1, JTC-801 -2, JTC-801-3 and JTC-801-4.
  • FIG. 1 1 shows graph showing inhibition of H4 cell proliferation in the presence of test compounds JTC-801-1, JTC-801-2, JTC-801-3 and JTC-801-4 compared to DMSO.
  • the Table summarizes the data.
  • FIG. 12 is a volcano plot showing significant captured hits applying compounds CPT- 753 (top) and CPT-754 (bottom) at 5 ⁇ in cell homogenate pellet fraction under optimized capture conditions.
  • FIG. 13 is a volcano plot showing significant captured hits applying compounds CPT- 753 (top) and CPT-754 (bottom) at 5 ⁇ in cell homogenate supernatant fraction under optimized capture conditions.
  • the present disclosure relates to methods of treating an mTOR-related disease, disorder or condition, and methods for treating proliferative diseases, disorders, and conditions using a 4-aminoquinoline compound identified herein as having cytotoxic and/or anti-proliferative activity.
  • the 4-aminoquinoline compound is referred to as JTC-801.
  • the biological activity of JTC-801 was discovered using a model system relevant to diseases and disorders characterized by inappropriate activation of the mTOR pathway (via deletion of the TSC2 gene).
  • JTC-801 exhibits enhanced cytotoxic activity in TSC2 null or deficient cells.
  • JTC-801 exhibits meaningful cytotoxic activity in other cancer cell lines not known to be dependent on constitutive activation of mTOR for their proliferation.
  • JTC-801 exhibits cytotoxic activity in mouse embryonic fibroblasts, angiomyolipomas, and various cancer cell lines, including HCT1 16 (colon) and 22RV1 (prostate).
  • JTC-801 selectively binds to two cellular enzymes, peroxisomal acyl-coenzyme A oxidase 3 (ACOX3) and peroxisomal acyl-coenzyme A oxidase 1 (ACOX1).
  • ACOX3 peroxisomal acyl-coenzyme A oxidase 3
  • ACOX1 peroxisomal acyl-coenzyme A oxidase 1
  • the disclosure also provides 4-aminoquinoline compounds for use in treating an mTOR-related disease, disorder or condition and for treating certain proliferative disorders, including cancers, and especially prostate cancer and chronic lymphocytic leukemia (CLL) that are not known to be mTOR-related.
  • CLL chronic lymphocytic leukemia
  • the disclosure also provides methods and related compositions for inhibiting cellular ACOX enzymes, especially ACOX1 and ACOX3.
  • JTC-801 N-(4-amino-2-methylquinoloin-6-yl)-2-[(4- ethylphenoxy)methyl]benzamide, and the CAS number is 244218-51-7.
  • JTC-801 is a 4-aminoquinoline derivative and this small molecule was initially investigated as a selective antagonist of the nociception receptor, also known as the ORL- 1 receptor.
  • ORL- 1 receptor The opioid like- 1 (ORL-1) receptor has a high homology with the opioid receptor but does not bind with conventional opioid ligands. It has been reported that the ORL-1 receptor plays a significant role in pathways of pain, anxiety, learning and memory, reward and tolerance, feeding, renal system and circadian rhythms.
  • JTC-801 has been investigated in numerous animal models of pain such as tail flick, hot plate, spinal nerve injury, inflammation, neuropathic pain, and allodynia. Clinical trials in humans for pain management have also been pursued (Lambert et al. Nature Reviews, 2008: 694-710).
  • the ORL- 1 receptor has been implicated in various disease states such as cardiovascular disease (congestive heart failure, heart failure, stroke, hypertension), neurogenic bladder, anxiety, drug abuse, depression, anorexia/obesity, Parkinson's disease, epilepsy, inflammation, neuroinflammation, immune system disorders and sepsis.
  • cardiovascular disease congestive heart failure, heart failure, stroke, hypertension
  • neurogenic bladder anxiety, drug abuse, depression, anorexia/obesity, Parkinson's disease, epilepsy, inflammation, neuroinflammation, immune system disorders and sepsis.
  • JTC-801 has not been reported to treat any of these diseases.
  • JTC-801 encompasses the preparation of 4,6-diamino-2- methylquinoline, which can be functionalized to obtain JTC-801 and derivatives thereof according to the methods described in U.S. Patent No. 6,410,561, and WO 1999048492, incorporated herein by reference.
  • the term "pharmaceutically acceptable salt,” is a salt formed from, for example, an acid and a basic group of a compound described herein (e.g., N-(4-amino-2- methylquinoloin-6-yl)-2-[(4-ethylphenoxy)methyl]benzamide).
  • Illustrative salts include, but are not limited, to sulfate, citrate, acetate, oxalate, chloride, bromide, iodide, nitrate, bisulfate, phosphate, acid phosphate, isonicotinate, lactate, salicylate, acid citrate, tartrate, oleate, tannate, pantothenate, bitartrate, ascorbate, succinate, maleate, besylate, gentisinate, fumarate, gluconate, glucaronate, saccharate, formate, benzoate, glutamate,
  • the salt of JTC-801 comprises monohydrochloride.
  • pharmaceutically acceptable salt also refers to a salt prepared from a compound described herein (e.g., N-(4-amino-2- methylquinoloin-6-yl)-2-[(4-ethylphenoxy)methyl]benzamide), having an acidic functional group, such as a carboxylic acid functional group, and a pharmaceutically acceptable inorganic or organic base.
  • a compound described herein e.g., N-(4-amino-2- methylquinoloin-6-yl)-2-[(4-ethylphenoxy)methyl]benzamide
  • Suitable bases include, but are not limited to, hydroxides of alkali metals such as sodium, potassium, and lithium; hydroxides of alkaline earth metal such as calcium and magnesium; hydroxides of other metals, such as aluminum and zinc; ammonia, and organic amines, such as unsubstituted or hydroxy-substituted mono-, di-, or trialkylamines;
  • dicyclohexylamine tributyl amine
  • pyridine N-methyl, N-ethylamine
  • diethylamine diethylamine
  • triethylamine mono-, bis-, or tris-(2-hydroxy-lower alkyl amines), such as mono-, bis-, or tris-(2-hydroxyethyl)amine, 2-hydroxy-tert-butylamine, or tris-(hydroxymethyl)methylamine, N, N,-di-lower alkyl-N-(hydroxy lower alkyl)-amines, such as N,N-dimethyl-N-(2- hydroxyethyl)amine, or tri-(2-hydroxyethyl)amine; N-methyl-D-glucamine; and amino acids such as arginine, lysine, and the like.
  • pharmaceutically acceptable salt also refers to a salt prepared from a compound described herein (e.g., N-(4-amino-2-methylquinoloin-6- yl)-2-[(4-ethylphenoxy)methyl]benzamide), having a basic functional group, such as an amino functional group, and a pharmaceutically acceptable inorganic or organic acid.
  • a compound described herein e.g., N-(4-amino-2-methylquinoloin-6- yl)-2-[(4-ethylphenoxy)methyl]benzamide
  • Suitable acids include hydrogen sulfate, citric acid, acetic acid, oxalic acid, hydrochloric acid (HC1), hydrogen bromide (HBr), hydrogen iodide (HI), nitric acid, hydrogen bisulfide, phosphoric acid, lactic acid, salicylic acid, tartaric acid, bitartratic acid, ascorbic acid, succinic acid, maleic acid, besylic acid, fumaric acid, gluconic acid, glucaronic acid, formic acid, benzoic acid, glutamic acid, methanesulfonic acid, ethanesulfonic acid, benzenesulfonic acid, and p-toluenesulfonic acid.
  • salts of the compounds described herein can be synthesized from the parent compound (e.g., N-(4-amino-2-methylquinoloin-6-yl)-2-[(4-ethylphenoxy)methyl]benzamide) by conventional chemical methods such as methods described in Pharmaceutical Salts:
  • salts can be prepared by reacting the parent compound (e.g., N-(4-amino-2-methylquinoloin-6-yl)-2-[(4- ethylphenoxy)methyl]benzamide) with the appropriate acid in water or in an organic solvent, or in a mixture of the two.
  • parent compound e.g., N-(4-amino-2-methylquinoloin-6-yl)-2-[(4- ethylphenoxy)methyl]benzamide
  • One salt form of a compound described herein can be converted to the free base and optionally to another salt form by methods well known to the skilled person.
  • the free base can be formed by passing the salt solution through a column containing an amine stationary phase (e.g. a Strata-NH 2 column).
  • a solution of the salt in water can be treated with sodium bicarbonate to decompose the salt and precipitate out the free base.
  • the free base may then be combined with another acid using routine methods.
  • polymorph means solid crystalline forms of a compound of the present disclosure (e.g., N-(4-amino-2-methylquinoloin-6-yl)-2-[(4- ethylphenoxy)methyl]benzamide) or complex thereof.
  • a compound of the present disclosure e.g., N-(4-amino-2-methylquinoloin-6-yl)-2-[(4- ethylphenoxy)methyl]benzamide
  • Different polymorphs of the same compound can exhibit different physical, chemical and/or spectroscopic properties. Different physical properties include, but are not limited to stability (e.g., to heat or light),
  • hydrate means a compound of the present disclosure (e.g., N-(4-amino-2-methylquinoloin-6-yl)-2-[(4-ethylphenoxy)methyl]benzamide) or a salt thereof, which further includes a stoichiometric or non-stoichiometric amount of water bound by non-covalent intermolecular forces.
  • clathrate means a compound of the present disclosure (e.g., N-(4-amino-2-methylquinoloin-6-yl)-2-[(4-ethylphenoxy)methyl]benzamide) or a salt thereof in the form of a crystal lattice that contains spaces (e.g., channels) that have a guest molecule (e.g., a solvent or water) trapped within.
  • a guest molecule e.g., a solvent or water
  • prodrug means a derivative of a compound described herein (e.g. , N-(4-amino-2-methylquinoloin-6-yl)-2- [(4-ethylphenoxy)methyl]benzamide) that can hydrolyze, oxidize, or otherwise react under biological conditions (in vitro or in vivo) to provide a compound of the disclosure.
  • Prodrugs may only become active upon such reaction under biological conditions, or they may have activity in their unreacted forms.
  • prodrugs contemplated in this disclosure include, but are not limited to, analogs or derivatives of a compound described herein (e.g., N-(4-amino-2-methylquinoloin-6-yl)-2- [(4-ethylphenoxy)methyl]benzamide) that comprise biohydrolyzable moieties such as biohydrolyzable amides, biohydrolyzable esters, biohydrolyzable carbamates,
  • prodrugs include derivatives of compounds of any one of the formulae disclosed herein that comprise -NO, -N0 2 , -ONO, or -ONO 2 moieties.
  • Prodrugs can typically be prepared using well-known methods, such as those described by Burger's Medicinal Chemistry and Drug Discovery (1995) 172-178, 949-982 (Manfred E. Wolff ed., 5th ed).
  • some of the compounds included in this disclosure have one or more double bonds, or one or more asymmetric centers.
  • Such compounds can occur as racemates, racemic mixtures, single enantiomers, individual diastereomers, diastereomeric mixtures, and cis- or trans- or E- or Z- double isomeric forms. All such isomeric forms of these compounds are expressly included in the present disclosure.
  • the compounds of this disclosure can also be represented in multiple tautomeric forms, in such instances, the disclosure expressly includes all tautomeric forms of the compounds described herein (e.g., alkylation of a ring system may result in alkylation at multiple sites, the disclosure expressly includes all such reaction products). All such isomeric forms of such compounds are expressly included in the present disclosure. All crystal forms of the compounds described herein (e.g., N-(4-amino-2- methylquinoloin-6-yl)-2-[(4-ethylphenoxy)methyl]benzamide) are expressly included in the present disclosure.
  • solvate or “pharmaceutically acceptable solvate,” is a solvate formed from the association of one or more solvent molecules to one of the compounds disclosed herein (e.g., N-(4-amino-2-methylquinoloin-6-yl)-2-[(4- ethylphenoxy)methyl]benzamide).
  • solvate includes hydrates (e.g., hemi-hydrate, mono-hydrate, dihydrate, trihydrate, tetrahydrate, and the like).
  • analog refers to a chemical compound that is structurally similar to another but differs slightly in composition (as in the replacement of one atom by an atom of a different element or in the presence of a particular functional group, or the replacement of one functional group by another functional group).
  • an analog is a compound that is similar or comparable in function and appearance, but not in structure or origin to the reference compound.
  • derivative refers to compounds that have a common core structure, and are substituted with various groups as described herein.
  • the compounds of the present disclosure can be combined with one or more additional therapeutically effective agents.
  • the therapeutic agent is an inhibitor of the mammalian target of rapamycin (mTOR) pathway.
  • mTOR inhibitors include but are not limited to mTOR inhibitors, e.g., rapamycin (also referred to as sirolimus), everolimus, temsirolimus, ridaforolimus, umirolimus, zotarolimus, AZD8055, ⁇ 128, WYE- 132, Torin- 1, pyrazolopyrimidine analogs PP242, PP30, PP487, PP121, KU0063794, KU-BMCL- 200908069-1, Wyeth-BMCL-200910075-9b, I ⁇ -128, XL388, AZD8055, P2281, and P529. See, e.g., Liu et al. Drug Disc. Today Ther. Strateg., 6(2): 47-55 (2009), incorporated herein by reference.
  • mTOR inhibitors useful in the disclosure include those compounds disclosed in US 2007/0112005, incorporated herein by reference, e.g., trans-4-[4-amino-5-(7- methoxy-lH-indol-2-yl)imidazo[5, l-f] [l,2,4]triazin-7-yl]cyclohexane carboxylic acid (also known as OSI-027), and any salts, solvates, hydrates, and other physical forms, crystalline or amorphous, thereof.
  • OSI-027 can be prepared according to US 2007/01 12005, incorporated herein by reference.
  • Another exemplary mTOR inhibitor is OXA-01. See, e.g., WO 2013/152342, incorporated herein by reference.
  • mTOR inhibitors include dual PI3K/mTOR inhibitors, e.g., PI- 103, GDC-094, LY294002, GDC0941, WAY-001, WYE-354, WAY-600, WYE-687, Wyeth-BMCL- 200910075- 16b, Wyeth-BMCL-200910096-27, KU0063794 and KUBMCL-200908069-5, BEZ235, NVP-BEZ235, XL-765, and GSK2126458. See, e.g., Liu et al. Drug Disc. Today Ther. Strateg., 6(2): 47-55 (2009), incorporated herein by reference.
  • Exemplary inhibitors of the mTOR pathway also include epigallocatechin gallate (EGCG), caffeine, curcumin, and resveratrol.
  • EGCG epigallocatechin gallate
  • caffeine caffeine
  • curcumin curcumin
  • resveratrol resveratrol
  • Additional agents ⁇ e.g., mTOR inhibitors) suitable for use in the disclosure include any prodrugs, derivatives, or analogs of the inhibitors disclosed herein.
  • further derivatives of rapamycin are known to the skilled person and include, for example, an O- substituted derivative in which the hydroxyl group on the cyclohexyl ring of rapamycin is replaced by— OR1, in which Rl is hydroxyalkyl, hydroxyalkoxyalkyl, acylaminoalkyl and aminoalkyl.
  • the mTOR pathway inhibitor can also be a polypeptide (e.g., an antibody or fragment thereof) or nucleic acid (e.g., a double-stranded small interfering RNA, a short hairpin RNA, a micro-RNA, an antisense oligonucleotide, a morpholino, a locked nucleic acid, or an aptamer) that binds to and inhibits the expression level or activity or a protein (or nucleic acid encoding the protein) in the mTOR pathway.
  • nucleic acid e.g., a double-stranded small interfering RNA, a short hairpin RNA, a micro-RNA, an antisense oligonucleotide, a morpholino, a locked nucleic acid, or an aptamer
  • the polypeptide or nucleic acid inhibits mTOR Complex 1 (mTORC l), regulatory-associated protein of mTOR (Raptor), mammalian lethal with SEC 13 protein 8 (MLST8), proline -rich Akt substrate of 40 kDa (PRAS40), DEP domain-containing mTOR-interacting protein (DEPTOR), mTOR Complex 2 (mTORC2), rapamycin-insensitive companion of mTOR (RICTOR), G protein beta subunit-like (G ⁇ L), mammalian stress-activated protein kinase interacting protein 1 (mSINl), paxillin, RhoA, Ras-related C3 botulinum toxin substrate 1 (Rac l), Cell division control protein 42 homolog (Cdc42), protein kinase C a (PKCa), the serine/threonine protein kinase Akt, phosphoinositide 3-kinase (PI3K), p70S6K, Ras, and/
  • an agent suitable for combination with a composition of the disclosure is a bronchodilator, e.g., theophylline, amphetamine, a short-acting ⁇ 2-agonist such as salbutamol, a long-acting ⁇ 2-agonist such as salmeterol or formoterol, or an anticholinergic such as tiotropium or ipratropium bromide.
  • a bronchodilator e.g., theophylline, amphetamine, a short-acting ⁇ 2-agonist such as salbutamol, a long-acting ⁇ 2-agonist such as salmeterol or formoterol, or an anticholinergic such as tiotropium or ipratropium bromide.
  • Other exemplary bronchodilators include levosalbutamol, pirbuterol, epinephrine, ephedrine, terbutaline, clenbuterol, bambuterol, and in
  • the therapeutic agent is being used in standard cancer therapy.
  • Standard cancer therapies include surgery (e.g., surgical removal of cancerous tissue), radiation therapy, hormonal therapy, anti-estrogen therapy, gene therapy, bone marrow transplantation, chemotherapeutic treatment, biological response modifier treatment, and certain combinations of the foregoing.
  • Radiation therapy includes, but is not limited to, x-rays or gamma rays that are delivered from either an externally applied source such as a beam, or by implantation of small radioactive sources.
  • the therapeutic agent is being used are chemotherapeutic agents.
  • Chemotherapeutic agents are non-peptidic (i.e., non-proteinaceous) compounds that reduce proliferation of cancer cells, and encompass cytotoxic agents and cytostatic agents.
  • Non- limiting examples of chemotherapeutic agents include alkylating agents, nitrosoureas, antimetabolites, antitumor antibiotics, plant (vinca) alkaloids, and steroid hormones.
  • Agents that act to reduce cellular proliferation are known in the art and widely used.
  • Such agents include alkylating agents, such as nitrogen mustards, nitrosoureas, ethylenimine derivatives, alkyl sulfonates, and triazenes, including, but not limited to, mechlorethamine, cyclophosphamide (Cytoxan®), melphalan (L-sarcolysin), carmustine (BCNU), lomustine (CCNU), semustine (methyl-CCNU), streptozocin, chlorozotocin, uracil mustard, chlormethine, ifosfamide, chlorambucil, pipobroman, triethylenemelamine,
  • alkylating agents such as nitrogen mustards, nitrosoureas, ethylenimine derivatives, alkyl sulfonates, and triazenes, including, but not limited to, mechlorethamine, cyclophosphamide (Cytoxan®), melphalan (L-sarcolysin),
  • triethylenethiophosphoramine busulfan, dacarbazine, and temozolomide.
  • Antimetabolite agents include folic acid analogs, pyrimidine analogs, purine analogs, and adenosine deaminase inhibitors, including, but not limited to, cytarabine (CYTOSAR-U), cytosine arabinoside, fluorouracil (5-FU), floxuridine (FudR), 6-thioguanine, 6- mercaptopurine (6-MP), pentostatin, 5-fluorouracil (5-FU), methotrexate, 10-propargyl-5,8- dideazafolate (PDDF, CB3717), 5,8-dideazatetrahydrofolic acid (DDATHF), leucovorin, fludarabine phosphate, pentostatine, and gemcitabine.
  • CYTOSAR-U cytarabine
  • cytosine arabinoside including, but not limited to, fluorouracil (5-FU), floxuridine (FudR), 6-thioguanine, 6-
  • Suitable natural products and their derivatives include, but are not limited to, Ara-C, paclitaxel (Taxol®), docetaxel (Taxotere®), deoxycoformycin, mitomycin-C, L- asparaginase, azathioprine; brequinar; alkaloids, e.g. vincristine, vinblastine, vinorelbine, vindesine, etc.; podophyllotoxins, e.g. etoposide, teniposide, etc.; antibiotics, e.g.
  • anthracycline daunorubicin hydrochloride (daunomycin, rubidomycin, cerubidine), idarubicin, doxorubicin, epirubicin and morpholino derivatives, etc.; phenoxizone biscyclopeptides, e.g. dactinomycin; basic glycopeptides, e.g. bleomycin; anthraquinone glycosides, e.g. plicamycin (mithramycin); anthracenediones, e.g. mitoxantrone;
  • azirinopyrrolo indolediones e.g. mitomycin
  • macrocyclic immunosuppressants e.g.
  • cyclosporine FK-506 (tacrolimus, prograf), rapamycin, etc.; and the like.
  • anti-proliferative cytotoxic agents are navelbene, CPT- 11, anastrazole, letrazole, capecitabine, reloxafine, cyclophosphamide, ifosamide, and droloxafine.
  • Microtubule affecting agents that have anti-proliferative activity are also suitable for use and include, but are not limited to, allocolchicine (NSC 406042), Halichondrin B (NSC 609395), colchicine (NSC 757), colchicine derivatives (e.g., NSC 33410), dolstatin 10 (NSC 376128), maytansine (NSC 153858), rhizoxin (NSC 332598), paclitaxel (Taxol®), Taxol® derivatives, docetaxel (Taxotere®), thiocolchicine (NSC 361792), trityl cysterin, vinblastine sulfate, vincristine sulfate, natural and synthetic epothilones including but not limited to, eopthilone A, epothilone B, discodermolide; estramustine, nocodazole, and the like.
  • chemotherapeutic agents include metal complexes, e.g. cisplatin (cis-DDP), carboplatin, etc.; ureas, e.g. hydroxyurea; and hydrazines, e.g. N-methylhydrazine;
  • epidophyllotoxin a topoisomerase inhibitor
  • procarbazine mitoxantrone
  • leucovorin tegafur
  • Other anti-proliferative agents of interest include immunosuppressants, e.g.
  • mycophenolic acid mycophenolic acid, thalidomide, desoxyspergualin, azasporine, leflunomide, mizoribine, azaspirane (SKF 105685); Iressa® (ZD 1839, 4-(3-chloro-4-fluorophenylamino)-7-methoxy- 6-(3-(4-morpholinyl)propoxy)qu- inazoline); etc.
  • Taxanes include paclitaxel, as well as any active taxane derivative or pro-drug.
  • Protaxel (which should be understood herein to include analogues, formulations, and derivatives such as, for example, docetaxel, TAXOL®, TAXOTE E® (a formulation of docetaxel), 10-desacetyl analogs of paclitaxel and 3N-desbenzoyl-3'N-t-butoxycarbonyl analogs of paclitaxel) may be readily prepared utilizing techniques known to those skilled in the art (see also WO 94/07882, WO 94/07881, WO 94/07880, WO 94/07876, WO 93/23555, WO 93/10076; U.S. Pat. Nos. 5,294,637; 5,283,253; 5,279,949; 5,274, 137; 5,202,448;
  • Paclitaxel should be understood to refer to not only the common chemically available form of paclitaxel, but analogs and derivatives (e.g., TAXOTERE® docetaxel, as noted above) and paclitaxel conjugates (e.g., paclitaxel-PEG, paclitaxel-dextran, or paclitaxel- xylose).
  • analogs and derivatives e.g., TAXOTERE® docetaxel, as noted above
  • paclitaxel conjugates e.g., paclitaxel-PEG, paclitaxel-dextran, or paclitaxel- xylose.
  • Taxane also included within the term “taxane” are a variety of known derivatives, including both hydrophilic derivatives, and hydrophobic derivatives. Taxane derivatives include, but not limited to, galactose and mannose derivatives described in International Patent
  • the therapeutic agent is a biological response modifier suitable for use in connection with the methods of the disclosure include, but are not limited to, (1) inhibitors of tyrosine kinase activity; (2) inhibitors of serine/threonine kinase activity; (3) tumor-associated antigen antagonists, such as antibodies that bind specifically to a tumor antigen; (4) apoptosis receptor agonists; (5) interleukin-2; (6) IFN-. alpha.; (7) IFN-. gamma.; (8) colony-stimulating factors; (9) inhibitors of angiogenesis; and (10) antagonists of tumor necrosis factor.
  • ABL, ACK, CSK, focal adhesion kinase (FAK), FES, FRK, JAK, SRC -A, SRC-B, TEC and SYK family are expressed, which can be subdivided in 19 families.
  • tyrosine kinases inhibitors include:
  • the SRC inhibitor which can be a polypeptide (e.g., an antibody or fragment thereof) or nucleic acid (e.g., a double-stranded small interfering RNA, a short hairpin RNA, a micro- RNA, an antisense oligonucleotide, a morpholino, a locked nucleic acid, or an aptamer) that binds to and inhibits the expression level or activity of the SRC protein or a nucleic acid encoding the SRC protein.
  • nucleic acid e.g., a double-stranded small interfering RNA, a short hairpin RNA, a micro- RNA, an antisense oligonucleotide, a morpholino, a locked nucleic acid, or an aptamer
  • the agent is a SRC inhibitor, e.g., bosutinib, saracatinib, dasatinib, ponatinib, KX2-391, XL-228, TG100435/TG100855, or DCC2036. See, e.g., Inc et al. Oncologist. 201 1 May; 16(5): 566-578, incorporated herein by reference.
  • the agent is a VEGF inhibitor (e.g., a VEGF-C or -D inhibitor) or a VEGF receptor inhibitor, e.g., bevacizumab, sunitinib, pazopanib, axitinib, sorafenib, regorafenib, lenvatinib, or motesanib.
  • a VEGF inhibitor e.g., a VEGF-C or -D inhibitor
  • a VEGF receptor inhibitor e.g., bevacizumab, sunitinib, pazopanib, axitinib, sorafenib, regorafenib, lenvatinib, or motesanib.
  • the VEGF inhibitor can be a polypeptide (e.g., an antibody or fragment thereof) or nucleic acid (e.g., a double-stranded small interfering RNA, a short hairpin RNA, a micro- RNA, an antisense oligonucleotide, a morpholino, a locked nucleic acid, or an aptamer) that binds to and inhibits the expression level or activity of a VEGF protein, a VEGF receptor protein, or a nucleic acid encoding one of these proteins.
  • the VEGF inhibitor is a soluble VEGF receptor (e.g., a soluble VEGF-C/D receptor (sVEGFR-3)).
  • the agent is a Janus kinase (JAK) inhibitor, e.g., tofacitinib, ruxolitinib, baricitinib, CYT387 (CAS number 1056634-68-4), lestaurtinib, pacritinib, or TG101348 (CAS number 936091-26-8).
  • JK Janus kinase
  • the JAK inhibitor can be a polypeptide (e.g., an antibody or fragment thereof) or nucleic acid (e.g., a double-stranded small interfering RNA, a short hairpin RNA, a micro- RNA, an antisense oligonucleotide, a morpholino, a locked nucleic acid, or an aptamer) that binds to and inhibits the expression level or activity of a JAK (e.g., JAK1, JAK2, JAK3, or TYK2) or a nucleic acid encoding the JAK protein.
  • a JAK e.g., JAK1, JAK2, JAK3, or TYK2
  • the agent is a Bcr-ABL ligand, e.g., imatinib mesylate (STI571, Gleevec), Nilotinib (AMN107), Tyrphostin AG1024, ON012380, Tyrphostin AG957, Adaphostin, NS-187 See, e.g Broekman et al WJCO 201 1 February 2 (2):80-93.
  • Bcr-ABL ligand e.g., imatinib mesylate (STI571, Gleevec), Nilotinib (AMN107), Tyrphostin AG1024, ON012380, Tyrphostin AG957, Adaphostin, NS-187 See, e.g Broekman et al WJCO 201 1 February 2 (2):80-93.
  • the agent can be a hormone therapy, e.g., an anti-estrogen therapy (e.g., an estrogen or estrogen receptor antagonist which reduces the level and/or activity of estrogen in the body), progestin and/or progesterone, megestrol acetate, androgen, or gonadotropin releasing hormone (GnRH) agonist.
  • an anti-estrogen therapy e.g., an estrogen or estrogen receptor antagonist which reduces the level and/or activity of estrogen in the body
  • progestin and/or progesterone megestrol acetate, androgen
  • gonadotropin releasing hormone (GnRH) agonist e.g., an anti-estrogen therapy
  • progestin and/or progesterone e.g., progestin and/or progesterone
  • megestrol acetate e.g., androgen
  • gonadotropin releasing hormone (GnRH) agonist e.g., an anti-est
  • the agent can be a statin (or HMG-CoA reductase inhibitor), e.g., atorvastatin, cerivastatin, fluvastatin, lovastatin, mevastatin, pitavastatin, pravastatin, rosuvastatin, or simvastatin.
  • statin or HMG-CoA reductase inhibitor
  • the statin is further used in combination with ezetimibe, niacin, or amlodipine.
  • the agent can be an autophagy inhibitor (e.g., chloroquine or hydroxychloroquine).
  • an autophagy inhibitor e.g., chloroquine or hydroxychloroquine.
  • the agent is doxycycline or tetracycline.
  • Hormone modulators and steroids that are suitable for use include, but are not limited to, adrenocorticosteroids, e.g. prednisone, dexamethasone, etc.; estrogens and pregestins, e.g. hydroxyprogesterone caproate, medroxyprogesterone acetate, megestrol acetate, estradiol, clomiphene, tamoxifen; etc.; and adrenocortical suppressants, e.g.
  • adrenocorticosteroids e.g. prednisone, dexamethasone, etc.
  • estrogens and pregestins e.g. hydroxyprogesterone caproate, medroxyprogesterone acetate, megestrol acetate, estradiol, clomiphene, tamoxifen; etc.
  • adrenocortical suppressants e.g.
  • estradiosteroids may inhibit T cell proliferation.
  • the agent can be an inhibitor of HSP90 (e.g., geldanamycin, 17-AAG or STA-9090 (ganetespib)).
  • HSP90 e.g., geldanamycin, 17-AAG or STA-9090 (ganetespib)
  • the agent can be an antibody targeting the immune checkpoint pathway that increases the body's immune system to fight cancer.
  • This includes antibodies targeting Programmed Death Receptor- 1 (PD1) (e.g., pembrolizumab (Keytruda®), or nivolumab (Opivo®)), antibodies targeting Programmed Death- Ligand- 1 (PD-Ll) (e.g., MPDL3280A) or antibodies targeting cytotoxic T-lymphocyte-associated protein-4 (CTLA-4) (e.g., Ipilimumab).
  • PD1 Programmed Death Receptor- 1
  • PD-Ll e.g., MPDL3280A
  • CTLA-4 cytotoxic T-lymphocyte-associated protein-4
  • JTC-801 as an antagonist of inappropriate or unregulated cell growth due to activation of the mTOR pathway. JTC-801 and related molecules are therefore likely to be useful in the treatment of diseases and disorders characterized by inappropriate mTOR activation, perhaps especially where mTOR activation is related to TSC2 deficiency. Such diseases and disorders are discussed below.
  • JTC-801 is not limited to TSC2 null or deficient cells, and instead is exhibited in other cells, including cancer cell lines, not known to be dependent on constitutive activation of mTOR for their proliferation.
  • JTC-801 was shown to exhibit cytotoxic activity in mouse embryonic fibroblasts, angiomyolipomas, and various cancer cell lines including HCT116 (colon) and 22RVI (prostate). JTC-801 and related molecules are therefore likely to be useful in the treatment of proliferative diseases and disorders not known to be driven by inappropriate mTOR activation. Such diseases and disorders are discussed below.
  • JTC-801 selectively binds peroxisomal acyl-coenzyme A oxidase 3 (ACOX3) and peroxisomal acyl-coenzyme A oxidase 1 (ACOXl).
  • ACOX3 peroxisomal acyl-coenzyme A oxidase 3
  • ACOXl peroxisomal acyl-coenzyme A oxidase 1
  • Beta-Oxidation of fatty acids by ACOX3 produces acetyl-CoA, which is a substrate in the citric acid cycle and therefore important in ATP-dependent metabolism. It has been shown that peroxisomal branched fatty acid beta-oxidation is upregulated in cancerous cells, e.g., prostate cancer (Zha et al, The Prostate 63:316-323 (2005)).
  • a study of citric acid cycle intermediates in benign and malignant prostatic homogenates showed that in early and advanced prostatic carcinoma reduction in citrate concentrations was directly proportional to the size and extent of the cancer (Cooper et al, JSR 3 (2), 1 12-121 (1963)).
  • ACOX3 has also been identified as a potential genetic marker in oncology applications for determining cancer identification and progression of chronic lymphocytic leukemia (Wade at el haematologica 96 (10), 1496-1503 (201 1)).
  • mTOR Diseases and Disorders have also been identified as a potential genetic marker in oncology applications for determining cancer identification and progression of chronic lymphocytic leukemia (Wade at el haematologica 96 (10), 1496-1503 (201 1)).
  • the mammalian target of rapamycin (mTOR) pathway is an important cellular signaling pathway that is involved in multiple physiological functions, ranging from cell growth, proliferation, and metabolism to protein synthesis and autophagy.
  • the mTOR protein is a 289 kDa serine -threonine kinase that is a member of the phosphoinositide 3- kinase (PI3K) -related kinase family.
  • mTOR forms two distinct complexes— mTOR complex 1 (mTORCl) and mTOR complex 2 (mTORC2).
  • the mTORCl and mTORC2 complexes have distinct sensitivities to rapamycin, upstream inputs, and downstream outputs.
  • the mTORC 1 pathway senses and integrates intracellular and extracellular signals, including growth factors, oxygen, amino acids, and energy status, in order to regulate a wide range of processes, such as protein and lipid synthesis and autophagy.
  • mTORC 1 is acutely sensitive to rapamycin. Rapamycin inhibits mTORC l by binding to FK506-binding protein of 12 kDa (FKBP12) and interacting with the FKBP12-rapamycin binding domain (FRB) of mTOR.
  • FKBP12 FK506-binding protein of 12 kDa
  • FKBP12 FKBP12-rapamycin binding domain
  • mTORC 1 When bound to GTP, Rheb interacts directly with mTORC l to stimulate its kinase activity. TSC1/TSC2 converts Rheb into its inactive GDP-bound form and is thus an inhibitor of mTORC 1.
  • mTORC 1 also acts to positively regulate ATP production.
  • mTORC l activates hypoxia inducible factor la (HIF 1 a), which positively regulates many glycolytic genes.
  • HIF 1 a hypoxia inducible factor la
  • mTORC 1 increases the expression of genes involved in oxidative metabolism and positively regulates mitochondrial biogenesis. Another mechanism by which mTORC l promotes growth is by negatively regulating autophagy, which is a degradative process in cells recycles damaged organelles and helps cells/organisms adapt to nutrient starvation.
  • mTOR pathway When the mTOR pathway is hyperactivated, increased cell growth and proliferation occurs.
  • Dysregulation of the mTOR pathway (such as by aberrant regulation of mTORC 1) is implicated in the pathophysiology of a number of diseases. These diseases include genetic tumor syndromes, neurological diseases, such as fragile X syndrome, Down syndrome, Rett syndrome, epilepsy, autism/neurodevelopmental disorders, and neurodegenerative diseases ⁇ e.g., Alzheimer's, Parkinson's, Huntington's disease, amyotrophic lateral sclerosis, and frontotemporal dementia). See, e.g., Wong. Biomed. J. 36.2(2013):40-50; and Dibble et al. Mol. Cell.
  • mTOR is thought to play a role in neurodegenerative diseases in part through its regulation of autophagy, which is essential for the survival of neural cells. Inhibition of mTORC 1 with rapamycin has been shown to promote degradation of aggregate -prone proteins through autophagy and reduce the severity of neurodegeneration in some in vivo models. See Laplante et al. Cell
  • Other diseases associated with the dysregulation of the mTOR pathway include metabolic diseases ⁇ e.g., obesity, diabetes such as Type I or Type II diabetes, lipotoxicity, hyperlipidemia, hyperglycemia, insulin resistance, hyperglycemia, hyperinsulinemia, hypoinsulinemic, fatty liver disease, or cirrhosis).
  • metabolic diseases ⁇ e.g., obesity, diabetes such as Type I or Type II diabetes, lipotoxicity, hyperlipidemia, hyperglycemia, insulin resistance, hyperglycemia, hyperinsulinemia, hypoinsulinemic, fatty liver disease, or cirrhosis.
  • mTORC l decreases food intake by increasing the expression in the hypothalamus of agouti-related peptide (AgRP) and orexigenic neuropeptide Y (NPY). Deregulation in mTORC 1 signaling has been suggested to play a role in the development of obesity.
  • AgRP agouti-related peptide
  • NPY orexigenic neuropeptide Y
  • mTORC l is a positive regulator of beta cell function, and activation of mTORC l causes a decrease in blood glucose, hyperinsulinemia, and improved glucose tolerance.
  • elevated mTORCl activity has been correlated with obesity in animals.
  • mTORC l is important for the proper regulation of beta cell mass and insulin secretion, over- activation of mTORC 1 can lead to insulin resistance. See Laplante et al. Cell
  • rapamycin mTOR pathway inhibitors ⁇ e.g., rapamycin
  • rapamycin has been shown to cause hyperlipidemia, severe insulin resistance, and glucose intolerance. See Laplante et al. Cell 149.2(2012):274-293.
  • dysregulation of the mTOR pathway leads to cancer.
  • Mutations in genes encoding proteins in the mTOR pathway ⁇ e.g., components of mTORC l or mTORC2, or proteins upstream and/or downstream of these complexes can lead to cancer.
  • loss of p53 which commonly occurs in cancer leads to hyperactivation of mTORC l .
  • Mutations arising in other proteins in the mTOR pathway can also lead to cancer cell proliferation and
  • tumorigenesis e.g., by promoting the expression of genes that regulate cell survival, cell cycle progression, angiogenesis, energy metabolism, and metastasis.
  • mTORC l is aberrantly regulated in the majority of sporadic cancers. See Dibble et al. Mol. Cell. 47 (2012):535-546.
  • mTOR dysregulation can cause proliferative disorders.
  • brain tumors such as gliomas (e.g., glioblastoma multiforme), sarcoma, breast cancer, lung cancer (e.g., non-small-cell lung cancer), mesothelioma, appendiceal cancer, genitourinary cancers (e.g., renal cell carcinoma, prostate, bladder, testicular, penile, cervical cancer, ovarian cancer, von Hippel Lindau disease), head and neck cancer, gastrointestinal tumors (e.g., hepatocellular carcinoma, gallbladder cancer, esophageal cancer, gastric cancer, colorectal cancer, or pancreatic cancer), neuroendocrine tumors (NETs), thyroid tumor, pituitary tumor, adrenal tumor, hematological malignancy (e.g., Non-Hodgkin's lymphoma, mantle cell lymphoma, myeloma, B-cell lymphoma, leukemia
  • rapamycin analog temsirolimus
  • FDA Food and Drug Administration
  • Everolimus is another rapamycin analog that has been approved for the treatment of tuberous sclerosis complex, which is a genetic disease caused by mutations in
  • TSC1/TSC2 Tuberous sclerosis complex patients develop non-malignant tumors in a number of organs, such as the brain. See Laplante et al. Cell 149.2(2012):274-293.
  • rapamycin analogs other small molecules have been developed that directly inhibit mTOR kinase activity, e.g., ATP-competitive inhibitors of mTOR, which block the phosphorylation of downstream targets of mTORC 1 and mTORC2.
  • ATP-competitive inhibitors of mTOR which block the phosphorylation of downstream targets of mTORC 1 and mTORC2.
  • the therapeutic efficacy of rapamycin analogs and mTOR kinase inhibitors in the treatment of cancer has been limited, likely because of the presence of negative feedback loops in the mTOR pathway.
  • dual PI3K/mTOR inhibitors compounds that inhibit both the mTOR and class I PI3K kinases, which have similar catalytic domain structures. These molecules inhibit mTORC l, mTORC2, and PI3K, thereby decreasing phosphorylation of Akt, S6K1, and 4E-BP1.
  • Phase I clinical trials with these dual inhibitors e.g., NVP-BEZ235 (Novartis) and XL-765 (Exelixis) have had promising efficacy in patients with various cancer types. See Laplante et al. Cell 149.2(2012):274-293.
  • mTOR signaling can lead to cardiovascular disorders, e.g., impaired angiogenesis, impaired tissue recovery and regeneration after ischemic/reperfusion injury.
  • cardiovascular disorders e.g., impaired angiogenesis, impaired tissue recovery and regeneration after ischemic/reperfusion injury.
  • hyperactivation of the mTOR pathway can also lead to cardiac/vascular dysfunction, e.g., cardiac failure. See, e.g., Chong et al. Trends Cardiovasc. Med. 21.5(201 1): 151-5.
  • tuberous sclerosis complex In the genetic disorder, tuberous sclerosis complex (TSC), hyperactivation of the mTOR pathway stimulates tumor growth in the brain and other organs.
  • Tuberous sclerosis, or tuberous sclerosis complex is a rare genetic disease that causes benign tumors to grow in the brain and other organs (e.g., the kidneys, heart, eyes, lungs, and skin).
  • TSC usually affects the central nervous system (CNS) and results in a number of symptoms, such as skin abnormalities, seizures, developmental delay, behavioral problems, and kidney disease.
  • TSC is caused by a mutation or defect in at least one of the two tumor suppressor genes, TSC 1 and TSC2.
  • the TSC1 gene produces a protein called hamartin.
  • the TSC2 gene produces a protein called tuberin.
  • the TSC 1 and TSC2 proteins act as growth suppressors by inhibiting mTOR.
  • the TSC 1 -TSC2 complex functions as a heterodimer and associates with the Tre2-Bub2-Cdcl6 (TBC) 1 domain family member 7 (TBC 1D7) protein to regulate mTOR signaling.
  • the TSC1-TSC2-TBC1D7 complex senses specific cellular growth conditions and inhibits the mTOR complex 1 (mTORC l) in order to limit cell growth in response to poor growth conditions.
  • mTORC 1 activity is stimulated by binding to the active, GTP-bound form of the small Ras- related GTPase Rheb (Ras homolog enriched in brain).
  • TSC1-TSC2-TBC1D7 complex inhibits mTORCl through its GTPase-activating protein (GAP) activity toward Rheb, i.e., TSC 1 -TSC2-TBC 1 D7 converts Rheb into its inactive GDP-bound state.
  • GAP GTPase-activating protein
  • TSC Tumors
  • seizures e.g., infantile spasms; tonic-clonic, tonic, akinetic, atypical absence, myoclonic, complex partial or generalized squires
  • learning disabilities mental retardation
  • behavior problems e.g., aggression, sudden rage, attention deficit hyperactivity disorder, acting out, obsessive-compulsive disorder, and repetitive, destructive, or self-harming behavior
  • autism and skin abnormalities.
  • tumors can grow in nearly any organ in TSC patients, they are more often found in the brain, kidneys, heart, lungs, and skin.
  • kidney disorders e.g., cysts and angiomyolipomas, which are benign growths containing fatty tissue and muscle cells. Cysts are usually small and in limited quantities, but in some individuals, a large number of cysts can also occur, e.g., in a pattern similar to polycystic kidney disease. In these patients, kidney function deteriorates, leading to kidney failure. In some instances, the cysts can bleed and lead to anemia. Although angiomyolipomas caused by TSC are usually asymptomatic, in some cases, they can grow to a large size that causes pain, kidney failure, and/or bleeding. Other kidney disorders that can develop from TSC (e.g., from an angiomyolipoma) include renal cell carcinoma and oncocytomas, which are benign tumors unique to individuals with TSC.
  • kidney disorders e.g., cysts and angiomyolipomas, which are benign growths containing fatty tissue and muscle cells.
  • the hearts of infants and young children suffering from TSC often contain tumors called cardiac rhabdomyomas. If these tumors are large or are in large numbers, they can block circulation and lead to death.
  • Another type of benign tumor sometimes found in TSC patients is a phakoma, which appear as white patches on the retina. Phakomas generally do not cause vision problems and are useful for diagnosing TSC.
  • additional tumors and cysts may be found in other areas of the body, such as the liver, lung, and pancreas, and can include bone cysts, rectal polyps, gum fibromas, and dental pits.
  • a number of skin abnormalities can occur in TSC patients, including for example, hypomelanic macules (lighter patches of skin that may appear anywhere on the body and are caused by a lack of melanin); reddish spots or bumps called facial
  • angiofibromas that appear on the face and contain blood vessels and fibrous tissue; raised, discolored areas on the forehead; areas of thick leathery, pebbly skin commonly found on the lower back or nape of the neck; small fleshy tumors called ungual or subungual fibromas that grow around and under the toenails or fingernails; molluscum fibrosum or skin tags that usually occur across the back of the neck and shoulders; flat brown marks; and poliosis (a patch of white hair that appears on the scalp or eyelids).
  • MMPH multinodular multifocal pneumocyte hyperplasia
  • Lymphangioleiomyomatosis is a rare disorder resulting from proliferation in the lung, kidney, and axial lymphatics of abnormal smooth muscle-like cells (LAM cells) that exhibit features of neoplasia and neural crest origin. Over time, these muscle cells form into bundles and grow into the walls of the airways, and blood and lymph vessels, causing them to become obstructed, blocking the flow of air, blood, and lymph vessels to and from the lungs, and preventing the lungs from providing oxygen to the rest of the body.
  • LAM cells smooth muscle-like cells
  • LAM can occur in the presence and absence of tuberous sclerosis complex (TSC- LAM) or sporadic (S-LAM), respectively. Cystic destruction of the lung with progressive pulmonary dysfunction and the presence of abdominal tumors (e.g., angiomyolipomas
  • lymphangioleiomyomas characterize the disease. LAM typically occurs in premenopausal women, suggesting the involvement of female hormones in disease pathogenesis.
  • LAM LAM-induced myeloma
  • PLCH pulmonary Langerhan's cell histiocytosis
  • lymphocytic interstitial pneumonitis Birtt Hogg Dube syndrome
  • Sjogren's syndrome Sjogren's syndrome
  • Rare syndromes of benign or malignant smooth muscle metastasis may also produce cystic change and closely mimic LAM, including benign metastasizing leiomyoma, endometrial stromal sarcomas, and low- grade leiomyosarcomas.
  • Classical methods of diagnosing LAM disease include chest X-rays, high-resolution CT scans and lung biopsies (e.g., thoracoscopy, open biopsy and
  • TSC or LAM There is currently no cure for TSC or LAM.
  • Various therapies are used to mitigate the symptoms and associated disorders.
  • antiepileptic drugs e.g., vigabatrin
  • SEGA brain tumors subependymal giant cell astrocytomas
  • SEGA brain tumors angiomyolipoma kidney tumors.
  • surgery is needed due to complications with tubers, subependymal nodules (SEN) or SEGA, or kidney tumors.
  • difficulty breathing due to LAM can be treated with supplemental oxygen therapy or lung transplantation, depending on the severity of the disease.
  • the present disclosure provides methods for the treatment of an mTOR disease, disorder or condition in a subject in need thereof by administering to the subject in need of such treatment, a therapeutically effective amount of a compound of the present disclosure, or a pharmaceutically acceptable salt, solvate, clathrate, hydrate, polymorph, metabolite, prodrug, analog or derivative thereof.
  • the present disclosure further provides the use of a compound of the present disclosure, or a pharmaceutically acceptable salt, solvate, clathrate, hydrate, polymorph, metabolite, prodrug, analog or derivative thereof, for the preparation of a medicament useful for the treatment of an mTOR disease, disorder or condition.
  • the present disclosure also provides methods of preventing an mTOR disease, disorder or condition in a subject in need thereof by administering a therapeutically effective amount of compound of the present disclosure, or a pharmaceutically acceptable salt, solvate, clathrate, hydrate, polymorph, metabolite, prodrug, analog or derivative thereof, to a subject in need of such treatment.
  • the present disclosure also provides the use of a compound of the present disclosure, or a pharmaceutically acceptable salt, solvate, clathrate, hydrate, polymorph, metabolite, prodrug, analog or derivative thereof, for the preparation of a medicament useful for the prevention of an mTOR disease, disorder or condition.
  • the mTOR disease, disorder or condition can be selected from genetic tumor syndromes, neurological diseases, fragile X syndrome, Down syndrome, Rett syndrome, epilepsy, autism/neurodevelopmental disorders, neurodegenerative diseases, metabolic diseases or anti-proliferative disorders, i.e., cancer.
  • the neurodegenerative disease can be Alzheimer's, Parkinson's, Huntington's disease, amyotrophic lateral sclerosis, or frontotemporal dementia.
  • the metabolic disease can be obesity, diabetes such as Type I or Type II diabetes, lipotoxicity, hyperlipidemia, hyperglycemia, insulin resistance, hyperglycemia,
  • hyperinsulinemia hyperinsulinemia, hypoinsulinemic, fatty liver disease, or cirrhosis.
  • compositions and methods for treating proliferative diseases and disorders that may not be based on aberrant mTOR activity.
  • Examples of such other proliferative disorders are cancers not known to rely on activated mTOR for proliferation and/or survival, including without limitation brain cancer, glioma, sarcoma, breast cancer, lung cancer, non-small-cell lung cancer, mesothelioma, appendiceal cancer, genitourinary cancers, renal cell carcinoma, prostate cancer, bladder cancer, testicular cancer, penile cancer, cervical cancer, ovarian cancer, von Hippel Lindau disease, head and neck cancer, gastrointestinal cancer, hepatocellular carcinoma, gallbladder cancer, esophageal cancer, gastric cancer, colorectal cancer, pancreatic cancer, neuroendocrine tumors, thyroid tumor, pituitary tumor, adrenal tumor, hematological malignancy, Hodgkin's lymphoma, Non-Hodgkin's lymphoma, mantle cell lymphoma, myeloma, B-cell lymphoma, leukemia and melanoma.
  • brain cancer
  • the disclosure provides a compound of Formula I, or a pharmaceutically acceptable salt, solvate, clathrate, hydrate, polymorph, metabolite, prodrug, analog or derivative thereof, for use in treating melanoma, breast cancer, prostate cancer, chronic myelogenous leukemia, or chronic lymphocyctic leukemia.
  • the disclosure provides a compound of Formula I for use in combination with at least one other active pharmaceutical agent (API) in the treatment of cancer.
  • the at least one other API is pazopanib, disulfiram, fulvestrant, or imatinib.
  • the present disclosure also provides methods for treating cancer by administering to a subject in need of such treatment a compound of Formula I, or a pharmaceutically acceptable salt, solvate, clathrate, hydrate, polymorph, metabolite, prodrug, analog or derivative thereof, or a pharmaceutical composition comprising same.
  • the cancer is melanoma, breast cancer, prostate cancer, chronic myelogenous leukemia, or chronic lymphocyctic leukemia.
  • the disclosure provides methods for treating cancer by administering to a subject in need of such treatment a compound of Formula I, or a pharmaceutically acceptable salt, solvate, clathrate, hydrate, polymorph, metabolite, prodrug, analog or derivative thereof, and a second API.
  • the second API is pazopanib, disulfiram, fulvestrant, or imatinib.
  • terapéuticaally effective amount refers to an amount of a compound of the present disclosure, or a pharmaceutically acceptable salt, solvate, clathrate, hydrate, polymorph, metabolite, prodrug, analog or derivative thereof, sufficient to treat, ameliorate a symptom of, reduce the severity of, or reduce the duration of an identified disease, disorder or condition, or enhance or improve the therapeutic effect of another therapy, or to prevent an identified disease, disorder or condition, or to exhibit a detectable therapeutic or inhibitory effect.
  • the effect can be detected by any assay method known in the art.
  • the precise effective amount for a subject will depend upon the subject's body weight, size, and health; the nature and extent of the condition; and the therapeutic or combination of therapeutics selected for administration.
  • the disease or condition to be treated is TSC and/or LAM and/or cancer.
  • the disease or condition to be treated is a TSC and/or LAM cell and /or cell proliferative disorder.
  • a therapeutically effective amount refers to the amount of a compound of the present disclosure, or a pharmaceutically acceptable salt, solvate, clathrate, hydrate, polymorph, metabolite, prodrug, analog or derivative thereof, that inhibits or reduces the proliferation of cancer and/or TSC and/or LAM cells, inhibits or reduces the spread of cancer and/or TSC and/or LAM cells (metastasis), or reduces the size of a tumor or improves FVC or FEV1 or reduces the amount of pleural effusion detectable by radiologic examination.
  • An effective amount of a compound of the present disclosure can be administered once daily, from two to five times daily, up to two times or up to three times daily, or up to eight times daily.
  • the compound of the present disclosure is administered once or twice daily, or less than once daily.
  • An effective amount of a compound of the present disclosure can range from about 0.001 mg/kg to about 1000 mg/kg, more preferably 0.01 mg/kg to about 100 mg/kg, more preferably 0.1 mg/kg to about 10 mg/kg; or any range in which the low end of the range is any amount between 0.001 mg/kg and 900 mg/kg and the upper end of the range is any amount between 0.1 mg/kg and 1000 mg/kg (e.g., 0.005 mg/kg and 200 mg/kg, 0.5 mg/kg and 20 mg/kg).
  • Effective doses will also vary, as recognized by those skilled in the art, depending on the diseases treated, route of administration, excipient usage, and the possibility of co-usage with other therapeutic treatments such as use of other agents. See, e.g., U.S. Patent No. 7,863,270, incorporated herein by reference.
  • the compound of the present disclosure e.g., JTC-801 or a pharmaceutically acceptable salt, solvate, clathrate, hydrate, polymorph, metabolite, prodrug, analog or derivative thereof
  • a dosage regimen of 30-100 mg/day e.g., 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, or 100 mg/day
  • 1 week e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 36, 48, or more weeks.
  • a dosage regimen of 100 mg/day for 4 or 8 weeks is administered at a dosage regimen of 30-100 mg/day (e.g., 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, or 100 mg/day) for at least 1 week (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 36, 48, or more weeks).
  • a dosage regimen of 100 mg/day for 4 or 8 weeks e.g., 1,
  • administered at a dosage regimen disclosed herein for 4-36 weeks ⁇ e.g., 4, 8, 12, 16, 24, or 36 weeks).
  • administered at a dosage regimen of 14 mg twice a day, 35 mg per day, 28 mg twice a day, 35 mg twice a day, or 70 mg per day e.g., for at least 1 week (e.g., 1, 2, 3, 4, or 5 weeks).
  • a dosage regimen disclosed herein for 4-36 weeks ⁇ e.g., 4, 8, 12, 16, 24, or 36 weeks.
  • administered at a dosage regimen of 21 mg twice a day, 35 mg per day, 35 mg twice a day, or 70 mg per day for 12 weeks See, e.g., Wada et al. PLoS One 7 (2012):e35069.
  • a "subject in need thereof is a subject having an mTOR disease, disorder or condition, or a subject having an increased risk of developing an mTOR disease, disorder or condition relative to the population at large.
  • the subject in need thereof is a subject having cancer and/or TSC and/or TAM or having an increased risk of developing cancer and/or TSC and/or LAM relative to the population at large.
  • the subject in need thereof can have ⁇ e.g., been diagnosed with) tuberous sclerosis complex (TSC) or the subject can have abnormal proliferation of LAM cells caused at least in part by an inactivating mutation in one of the tuberous sclerosis complex tumor suppressor genes, TSC l or TSC2 or the subject in need thereof can have cancer.
  • TSC tuberous sclerosis complex
  • the subject in need thereof can have ⁇ e.g., been diagnosed with) sporadic LAM, such that, the subject may not have clinical manifestations of TSC, nor have germline mutations in the TSC l or TSC2 tumor suppressor genes.
  • a subject with an increased risk of developing TSC and/or LAM relative to the population at large is a female subject with a family history or personal history of TSC and/or LAM.
  • a subject with an increased risk of developing TSC and/or relative to the population at large is a female subject having a germ-line or spontaneous mutation in TSC l or TSC2, or both.
  • a subject with an increased risk of developing TSC and/or relative to the population at large is a female subject with a family history of TSC and/or LAM and a germ-line or spontaneous mutation in TSC 1 or TSC2, or both.
  • the subject in need thereof can be one that is "non-responsive” or “refractory” to a currently available therapy for cancer and/or TSC and/or LAM.
  • the terms "non-responsive” and “refractory” refer to the subject's response to therapy as not clinically adequate to relieve one or more symptoms associated with cancer and/or TSC and/or LAM.
  • the mammal can be e.g., any mammal, e.g., a human, primate, bird, mouse, rat, fowl, dog, cat, cow, horse, goat, camel, sheep or a pig.
  • the mammal is a human.
  • the terms "subject” and "patient” are used
  • the present disclosure provides a monotherapy for the treatment of a disease, disorder or condition of the present disclosure.
  • monotherapy refers to the administration of a single active or therapeutic compound to a subject in need thereof.
  • monotherapy will involve administration of a therapeutically effective amount of an active compound.
  • monotherapy with one of the compound of the present disclosure, or a pharmaceutically acceptable salt, solvate, clathrate, hydrate, polymorph, metabolite, prodrug, analog or derivative thereof, to a subject in need of treatment.
  • Monotherapy may be contrasted with combination therapy, in which a combination of multiple active compounds is administered, preferably with each component of the combination present in a therapeutically effective amount.
  • treatment describes the management and care of a patient for the purpose of combating a disease, condition, or disorder and includes the administration of a compound of the present disclosure, or a pharmaceutically acceptable salt, solvate, clathrate, hydrate, polymorph, metabolite, prodrug, analog or derivative thereof, to alleviate the symptoms or complications of a disease, condition or disorder, or to eliminate the disease, condition or disorder.
  • FVC or FEV1 may refer to improvement in FVC or FEV1, reduction in the amount of pleural effusion detectable by radiologic examination, reduction in proliferation of cancer and/or TSC and/or LAM cells, reduction in the spread (metastasis) of cancer and/or TSC and/or LAM cells, reduction in the progression of a cancer and/or TSC and/or LAM-associated cancer, reduction in the size of a TSC and/or LAM-associated tumor and/or non-TSC and/or LAM-associated tumor, or reduction in the involvement of axial lymphatics.
  • prevention describes reducing or eliminating the onset of the symptoms or complications of the disease, condition or disorder and includes the administration of a compound of the present disclosure, or a
  • the term "alleviate” is meant to describe a process by which the severity of a sign or symptom of a disorder is decreased.
  • a sign or symptom can be alleviated without being eliminated.
  • the administration of compound of the disclosure leads to the elimination of a sign or symptom, however, elimination is not required.
  • Effective dosages are expected to decrease the severity of a sign or symptom.
  • a sign or symptom of a disorder such as cancer and/or TSC and/or LAM, which can occur in multiple locations, is alleviated if the severity of the cancer and/or TSC and/or LAM is decreased within at least one of multiple locations.
  • severity is meant to describe the transformation from abnormally proliferating smooth-muscle like cells in the lungs to the formation of cysts, to the formation of tumors.
  • severity describes the degree to which a non-TSC and/or LAM-derived tumor and/or a TSC and/or LAM-derived tumor has secreted growth factors, degraded the extracellular matrix, become vascularized, lost adhesion to juxtaposed tissues, or metastasized.
  • severity describes the number of locations to which a tumor has metastasized.
  • severity includes the difficulty of treating tumors of varying types and locations.
  • symptom is defined as an indication of disease, illness, injury, or that something is not right in the body. Symptoms are felt or noticed by the individual experiencing the symptom, but may not easily be noticed by others. Others are defined as non-health-care professionals.
  • signs are also defined as an indication that something is not right in the body. But signs are defined as things that can be seen by a doctor, nurse, or other health care professional.
  • Treating a disorder, disease or condition of the present disclosure can result in a reduction in size of a tumor.
  • a reduction in size of a tumor may also be referred to as "tumor regression".
  • tumor size is reduced by 5% or greater relative to its size prior to treatment; more preferably, tumor size is reduced by 10% or greater; more preferably, reduced by 20% or greater; more preferably, reduced by 30% or greater; more preferably, reduced by 40% or greater; even more preferably, reduced by 50% or greater; and most preferably, reduced by greater than 75% or greater.
  • Size of a tumor may be measured by any reproducible means of measurement. The size of a tumor may be measured as a diameter of the tumor.
  • Treating a disorder, disease or condition of the present disclosure can result in a reduction in tumor volume.
  • tumor volume is reduced by 5% or greater relative to its size prior to treatment; more preferably, tumor volume is reduced by 10% or greater; more preferably, reduced by 20% or greater; more preferably, reduced by 30% or greater; more preferably, reduced by 40% or greater; even more preferably, reduced by 50% or greater; and most preferably, reduced by greater than 75% or greater.
  • Tumor volume may be measured by any reproducible means of measurement.
  • Treating a disorder, disease or condition of the present disclosure can result in a decrease in number of tumors.
  • tumor number is reduced by 5% or greater relative to number prior to treatment; more preferably, tumor number is reduced by 10% or greater; more preferably, reduced by 20% or greater; more preferably, reduced by 30% or greater; more preferably, reduced by 40% or greater; even more preferably, reduced by 50% or greater; and most preferably, reduced by greater than 75%.
  • Number of tumors may be measured by any reproducible means of measurement. The number of tumors may be measured by counting tumors visible to the naked eye or at a specified magnification.
  • the specified magnification is 2x, 3x, 4x, 5x, lOx, or 50x.
  • Treating a disorder, disease or condition of the present disclosure can result in a decrease in number of metastatic lesions in other tissues or organs distant from the primary tumor site.
  • the number of metastatic lesions is reduced by 5% or greater relative to number prior to treatment; more preferably, the number of metastatic lesions is reduced by 10% or greater; more preferably, reduced by 20% or greater; more preferably, reduced by 30% or greater; more preferably, reduced by 40% or greater; even more preferably, reduced by 50% or greater; and most preferably, reduced by greater than 75%.
  • the number of metastatic lesions may be measured by any reproducible means of measurement.
  • the number of metastatic lesions may be measured by counting metastatic lesions visible to the naked eye or at a specified magnification.
  • the specified magnification is 2x, 3x, 4x, 5x, lOx, or 50x.
  • Treating a disorder, disease or condition of the present disclosure can result in a decrease in amount of pleural effusion that accumulates in the lungs.
  • the amount of pleural effusion is reduced by 5% or greater relative to number prior to treatment; more preferably, the amount of pleural effusion is reduced by 10% or greater; more preferably, reduced by 20% or greater; more preferably, reduced by 30% or greater; more preferably, reduced by 40% or greater; even more preferably, reduced by 50% or greater; and most preferably, reduced by greater than 75%.
  • the amount of pleural effusion may be measured by any reproducible means of measurement.
  • Treating a disorder, disease or condition of the present disclosure can result in an increase in average survival time of a population of treated subjects in comparison to a population receiving carrier alone.
  • the average survival time is increased by more than 30 days; more preferably, by more than 60 days; more preferably, by more than 90 days; and most preferably, by more than 120 days.
  • An increase in average survival time of a population may be measured by any reproducible means.
  • An increase in average survival time of a population may be measured, for example, by calculating for a population the average length of survival following initiation of treatment with an active compound.
  • An increase in average survival time of a population may also be measured, for example, by calculating for a population the average length of survival following completion of a first round of treatment with an active compound.
  • Treating a disorder, disease or condition of the present disclosure can result in an increase in average survival time of a population of treated subjects in comparison to a population of untreated subjects.
  • the average survival time is increased by more than 30 days; more preferably, by more than 60 days; more preferably, by more than 90 days; and most preferably, by more than 120 days.
  • An increase in average survival time of a population may be measured by any reproducible means.
  • An increase in average survival time of a population may be measured, for example, by calculating for a population the average length of survival following initiation of treatment with an active compound.
  • An increase in average survival time of a population may also be measured, for example, by calculating for a population the average length of survival following completion of a first round of treatment with an active compound.
  • Treating a disorder, disease or condition of the present disclosure can result in increase in average survival time of a population of treated subjects in comparison to a population receiving monotherapy with a drug that is not a compound of the present disclosure, or a pharmaceutically acceptable salt, solvate, clathrate, hydrate, polymorph, metabolite, prodrug, analog or derivative thereof.
  • the average survival time is increased by more than 30 days; more preferably, by more than 60 days; more preferably, by more than 90 days; and most preferably, by more than 120 days.
  • An increase in average survival time of a population may be measured by any reproducible means.
  • An increase in average survival time of a population may be measured, for example, by calculating for a population the average length of survival following initiation of treatment with an active compound.
  • An increase in average survival time of a population may also be measured, for example, by calculating for a population the average length of survival following completion of a first round of treatment with an active compound.
  • Treating a disorder, disease or condition of the present disclosure can result in a decrease in the mortality rate of a population of treated subjects in comparison to a population receiving carrier alone. Treating a disorder, disease or condition of the present disclosure can result in a decrease in the mortality rate of a population of treated subjects in comparison to an untreated population. Treating a disorder, disease or condition of the present disclosure can result in a decrease in the mortality rate of a population of treated subjects in comparison to a population receiving monotherapy with a drug that is not a compound of the present disclosure, or a pharmaceutically acceptable salt, solvate, clathrate, hydrate, polymorph, metabolite, prodrug, analog or derivative thereof.
  • the mortality rate is decreased by more than 2%; more preferably, by more than 5%; more preferably, by more than 10%; and most preferably, by more than 25%.
  • a decrease in the mortality rate of a population of treated subjects may be measured by any reproducible means.
  • a decrease in the mortality rate of a population may be measured, for example, by calculating for a population the average number of disease-related deaths per unit time following initiation of treatment with an active compound.
  • a decrease in the mortality rate of a population may also be measured, for example, by calculating for a population the average number of disease-related deaths per unit time following completion of a first round of treatment with an active compound.
  • Treating a disorder, disease or condition of the present disclosure can result in a decrease in tumor growth rate.
  • tumor growth rate is reduced by at least 5% relative to number prior to treatment; more preferably, tumor growth rate is reduced by at least 10%; more preferably, reduced by at least 20%; more preferably, reduced by at least 30%; more preferably, reduced by at least 40%; more preferably, reduced by at least 50%; even more preferably, reduced by at least 50%; and most preferably, reduced by at least 75%.
  • Tumor growth rate may be measured by any reproducible means of measurement. Tumor growth rate can be measured according to a change in tumor diameter per unit time.
  • Treating a disorder, disease or condition of the present disclosure can result in a decrease in tumor regrowth.
  • tumor regrowth is less than 5%; more preferably, tumor regrowth is less than 10%; more preferably, less than 20%; more preferably, less than 30%; more preferably, less than 40%; more preferably, less than 50%; even more preferably, less than 50%; and most preferably, less than 75%.
  • Tumor regrowth may be measured by any reproducible means of measurement. Tumor regrowth is measured, for example, by measuring an increase in the diameter of a tumor after a prior tumor shrinkage that followed treatment. A decrease in tumor regrowth is indicated by failure of tumors to reoccur after treatment has stopped.
  • Treating or preventing a cell proliferative disorder of the present disclosure can result in a reduction in the rate of cellular proliferation.
  • the rate of cellular proliferation is reduced by at least 5%; more preferably, by at least 10%; more preferably, by at least 20%; more preferably, by at least 30%; more preferably, by at least 40%; more preferably, by at least 50%; even more preferably, by at least 50%; and most preferably, by at least 75%.
  • the rate of cellular proliferation may be measured by any reproducible means of measurement.
  • the rate of cellular proliferation is measured, for example, by measuring the number of dividing cells in a tissue sample per unit time.
  • Treating or preventing a cell proliferative disorder of the present disclosure can result in a reduction in the proportion of proliferating cells.
  • the proportion of proliferating cells is reduced by at least 5%; more preferably, by at least 10%; more preferably, by at least 20%; more preferably, by at least 30%; more preferably, by at least 40%; more preferably, by at least 50%; even more preferably, by at least 50%; and most preferably, by at least 75%.
  • the proportion of proliferating cells may be measured by any reproducible means of measurement.
  • the proportion of proliferating cells is measured, for example, by quantifying the number of dividing cells relative to the number of nondividing cells in a tissue sample.
  • the proportion of proliferating cells can be equivalent to the mitotic index.
  • Treating or preventing a cell proliferative disorder of the present disclosure can result in a decrease in size of an area or zone of cellular proliferation.
  • size of an area or zone of cellular proliferation is reduced by at least 5% relative to its size prior to treatment; more preferably, reduced by at least 10%; more preferably, reduced by at least 20%; more preferably, reduced by at least 30%; more preferably, reduced by at least 40%; more preferably, reduced by at least 50%; even more preferably, reduced by at least 50%; and most preferably, reduced by at least 75%.
  • Size of an area or zone of cellular proliferation may be measured by any reproducible means of measurement.
  • the size of an area or zone of cellular proliferation may be measured as a diameter or width of an area or zone of cellular proliferation.
  • Treating or preventing a cell proliferative disorder of the present disclosure can result in a decrease in the number or proportion of cells having an abnormal appearance or morphology.
  • the number of cells having an abnormal morphology is reduced by at least 5% relative to its size prior to treatment; more preferably, reduced by at least 10%; more preferably, reduced by at least 20%; more preferably, reduced by at least 30%; more preferably, reduced by at least 40%; more preferably, reduced by at least 50%; even more preferably, reduced by at least 50%; and most preferably, reduced by at least 75%.
  • An abnormal cellular appearance or morphology may be measured by any reproducible means of measurement.
  • An abnormal cellular morphology can be measured by microscopy, e.g., using an inverted tissue culture microscope.
  • An abnormal cellular morphology can take the form of nuclear pleiomorphism.
  • the term "selectively" means tending to occur at a higher frequency in one population than in another population.
  • the compared populations can be cell populations.
  • a compound of the present disclosure, or a pharmaceutically acceptable salt, solvate, clathrate, hydrate, polymorph, metabolite, prodrug, analog or derivative thereof acts selectively on a hyper-proliferating cells but not on a normal cell.
  • a "normal cell” is a cell that cannot be classified as part of a "cell proliferative disorder".
  • a normal cell lacks unregulated or abnormal growth, or both, that can lead to the development of an unwanted condition or disease.
  • a normal cell possesses normally functioning cell cycle checkpoint control mechanisms.
  • a compound of the present disclosure acts selectively to modulate one molecular target (e.g., a target kinase) but does not significantly modulate another molecular target (e.g., a non-target kinase).
  • the disclosure also provides a method for selectively inhibiting the activity of an enzyme, such as a kinase.
  • an event occurs selectively in population A relative to population B if it occurs greater than two times more frequently in population A as compared to population B.
  • An event occurs selectively if it occurs greater than five times more frequently in population A.
  • An event occurs selectively if it occurs greater than ten times more frequently in population A; more preferably, greater than fifty times; even more preferably, greater than 100 times; and most preferably, greater than 1000 times more frequently in population A as compared to population B.
  • cell death would be said to occur selectively in diseased or hyper-proliferating cells if it occurred greater than twice as frequently in diseased or hyper-proliferating cells as compared to normal cells.
  • the present disclosure also provides a combination therapy for the treatment of a disorder, disease or condition of the present disclosure.
  • “combination therapy” or “co-therapy” includes the administration of a compound of the present disclosure, or a pharmaceutically acceptable salt, solvate, clathrate, hydrate, polymorph, metabolite, prodrug, analog or derivative thereof, and at least a second agent, as disclosed herein, as part of a specific treatment regimen intended to provide the beneficial effect from the co-action of these therapeutic compounds.
  • the beneficial effect of the combination includes, but is not limited to, pharmacokinetic or pharmacodynamic co-action resulting from the combination of therapeutic compounds.
  • “Combination therapy” may be, but generally is not, intended to encompass the administration of two or more of these therapeutic compounds as part of separate monotherapy regimens that incidentally and arbitrarily result in the combinations of the present disclosure.
  • a compound of the present disclosure is administered in combination with one or more additional agents
  • administration may be simultaneous with or sequential to the administration of the one or more additional agents.
  • the one or more additional agents may be administered prior to (e.g., 5 minutes, 15 minutes, 30 minutes, 45 minutes, 1 hour, 2 hours, 4 hours, 6 hours, 12 hours, 24 hours, 48 hours, 72 hours, 96 hours, 1 week, 2 weeks, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 8 weeks, or 12 weeks before), concomitantly with, or subsequent to (e.g., 5 minutes, 15 minutes, 30 minutes, 45 minutes, 1 hour, 2 hours, 4 hours, 6 hours, 12 hours, 24 hours, 48 hours, 72 hours, 96 hours, 1 week, 2 weeks, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 8 weeks, or 12 weeks after) the administration of a compound of the present disclosure.
  • the methods described here incorporate the use of an additional pharmaceutically active agent (API), in addition to the 4-aminoquinoline compound, either in the same or different dosage forms.
  • API additional pharmaceutically active agent
  • the 4-aminoquinoline compound and the additional API exhibit synergistic activity.
  • the additional agent can be formulated for co-administration with a compound of the present disclosure in a single dosage form, as described in greater detail herein.
  • the additional agent can be administered separately from the dosage form that comprises the compound of the present disclosure.
  • the additional agent can be by the same or a different route of administration as the compound of the instant disclosure.
  • the administration of a composition comprising a compound of the present disclosure in combination with one or more additional agents provides a synergistic response in the subject having a disorder, disease or condition of the present disclosure.
  • the term "synergistic” refers to the efficacy of the combination being more effective than the additive effects of either single therapy alone.
  • the synergistic effect of combination therapy according to the disclosure can permit the use of lower dosages and/or less frequent administration of at least one agent in the combination compared to its dose and/or frequency outside of the combination.
  • the synergistic effect can be manifested in the avoidance or reduction of adverse or unwanted side effects associated with the use of either therapy in the combination alone.
  • a compound of the present disclosure is administered with a tyrosine kinase inhibitor, as described herein. More preferably, the tyrosine kinase inhibitor is pazopanib and/or imatinib.
  • Pazopanib and/or imatinib can be administered orally.
  • Pazopanib and/or imatinib can be administered in the form of capsules or tablets.
  • the administered dose of pazopanib and/or imatinib can be from 10 to 100 nanograms, from 50 to 250 nanograms, from 100 to 500 nanograms, from 500 to 1000 nanograms (0.5 to 1 microgram), or from 1000 to 2000 nanograms (1 to 2 micrograms).
  • the pazopanib and/or imatinib can be administered once daily, from two to five times daily, up to two times or up to three times daily, or up to eight times daily.
  • the sirolimus is administered once or twice daily, or less than once daily.
  • the total daily dose of pazopanib and/or imatinib administered to the subject can be 10 to 100 nanograms, from 50 to 250 nanograms, from 100 to 500 nanograms, from 500 to 1000 nanograms (0.5 to 1 microgram), or from 1000 to 2000 nanograms (1 to 2 micrograms).
  • Combination therapy also embraces the administration of the compounds of the present invenion in further combination with non-drug therapies (e.g., surgery or radiation treatment).
  • the combination therapy further comprises a non-drug treatment
  • the non- drug treatment may be conducted at any suitable time so long as a beneficial effect from the co-action of the combination of the therapeutic compounds and non-drug treatment is achieved.
  • the beneficial effect is still achieved when the non-drug treatment is temporally removed from the administration of the therapeutic compounds, perhaps by days or even weeks.
  • the non-drug treatment can be selected from chemotherapy, radiation therapy, hormonal therapy, anti-estrogen therapy, gene therapy, bone marrow transplantation, biological response modifier treatment and surgery.
  • a non-drug therapy is the removal of an ovary (e.g., to reduce the level of estrogen in the body), thoracentesis (e.g., to remove fluid from the chest), paracentesis (e.g., to remove fluid from the abdomen), surgery to remove or shrink angiomyolipomas, lung transplantation (and optionally with an antibiotic to prevent infection due to transplantation), or oxygen therapy (e.g., through a nasal cannula containing two small plastic tubes or prongs that are placed in both nostrils, through a face mask that fits over the nose and mouth, or through a small tube inserted into the windpipe through the front of the neck, also called transtracheal oxygen therapy).
  • ovary e.g., to reduce the level of estrogen in the body
  • thoracentesis e.g
  • compositions comprising a compound of the present disclosure, or a pharmaceutically acceptable salt, solvate, clathrate, hydrate, polymorph, metabolite, prodrug, analog or derivative thereof, in combination with at least one pharmaceutically acceptable excipient or carrier.
  • a "pharmaceutical composition” is a formulation containing the compounds of the present disclosure in a pharmaceutically acceptable form suitable for administration to a subject.
  • pharmaceutically acceptable refers to those compounds, materials, compositions, carriers, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
  • “Pharmaceutically acceptable excipient” means an excipient that is useful in preparing a pharmaceutical composition that is generally safe, non-toxic and neither biologically nor otherwise undesirable, and includes excipient that is acceptable for veterinary use as well as human pharmaceutical use.
  • pharmaceutically acceptable excipients include, without limitation, sterile liquids, water, buffered saline, ethanol, polyol (for example, glycerol, propylene glycol, liquid polyethylene glycol and the like), oils, detergents, suspending agents, carbohydrates (e.g., glucose, lactose, sucrose or dextran), antioxidants (e.g., ascorbic acid or glutathione), chelating agents, low molecular weight proteins, or suitable mixtures thereof.
  • compositions comprising the compounds of the present disclosure may be manufactured in a manner that is generally known, e.g., by means of conventional mixing, dissolving, granulating, dragee-making, levigating, emulsifying, encapsulating, entrapping, or lyophilizing processes. Techniques for formulation and administration of the disclosed compounds and pharmaceutical compositions of the disclosure can be found in Remington: the Science and Practice of Pharmacy, 19 th edition, Mack Publishing Co., Easton, PA (1995). [175] A pharmaceutical composition of the present disclosure can be provided in bulk or in dosage unit form. It is especially advantageous to formulate pharmaceutical compositions in dosage unit form for ease of administration and uniformity of dosage.
  • dosage unit form refers to physically discrete units suited as unitary dosages for the subject to be treated; each unit containing a predetermined quantity of active compound calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier.
  • the specification for the dosage unit forms of the disclosure are dictated by and directly dependent on the unique characteristics of the active compound and the particular therapeutic effect to be achieved.
  • a dosage unit form can be an ampoule, a vial, a suppository, a dragee, a tablet, a capsule, an IV bag, or a single pump on an aerosol inhaler.
  • the dosages of the pharmaceutical compositions used in accordance with the disclosure vary depending on the agent, the age, weight, and clinical condition of the recipient patient, and the experience and judgment of the clinician or practitioner administering the therapy, among other factors affecting the selected dosage.
  • the dose should be a therapeutically effective amount.
  • Dosages can be provided in mg/kg/day units of measurement (which dose may be adjusted for the patient's weight in kg, body surface area in m 2 , and age in years).
  • An effective amount of a pharmaceutical composition is that which provides an objectively identifiable improvement as noted by the clinician or other qualified observer. For example, alleviating a symptom of a disorder, disease or condition.
  • the term "dosage effective manner" refers to amount of a pharmaceutical composition to produce the desired biological effect in a subject or cell.
  • the dosage unit form can comprise 1 nanogram to 2 milligrams, or 0.1 milligrams to 2 grams; or from 10 milligrams to 1 gram, or from 50 milligrams to 500 milligrams or from 1 microgram to 20 milligrams; or from 1 microgram to 10 milligrams; or from 0.1 milligrams to 2 milligrams of a compound of the present disclosure.
  • compositions of the disclosure can take any suitable form (e.g, liquids, aerosols, solutions, inhalants, mists, sprays; or solids, powders, ointments, pastes, creams, lotions, gels, patches and the like) for administration by any desired route (e.g, pulmonary, inhalation, intranasal, oral, buccal, sublingual, parenteral, subcutaneous, intravenous, intramuscular, intraperitoneal, intrapleural, intrathecal, transdermal, transmucosal, rectal, and the like).
  • suitable form e.g, liquids, aerosols, solutions, inhalants, mists, sprays; or solids, powders, ointments, pastes, creams, lotions, gels, patches and the like
  • any desired route e.g, pulmonary, inhalation, intranasal, oral, buccal, sublingual, parenteral, subcutaneous, intravenous, intramuscular, intraperitoneal, intraple
  • a pharmaceutical composition of the disclosure may be in the form of an aqueous solution or powder for aerosol administration by inhalation or insufflation (either through the mouth or the nose), in the form of a tablet or capsule for oral administration; in the form of a sterile aqueous solution or dispersion suitable for administration by either direct injection or by addition to sterile infusion fluids for intravenous infusion; or in the form of a lotion, cream, foam, patch, suspension, solution, or suppository for transdermal or transmucosal administration.
  • a pharmaceutical composition of the disclosure can be in the form of an aerosol formulation suitable for pulmonary administration. Pulmonary administration is
  • compositions of the disclosure comprise particles of a compound of the present disclosure, or particles comprising a compound of the present disclosure, that are of a size range suitable for delivery to the lung.
  • the particles will have an average mean diameter of less than 10 microns or less than 5 microns, and preferably between 0.5 and 6 microns for deep lung delivery.
  • a particle size range of from about 2 to 10 microns, from about 5 to 10 microns or from about 5 to 15 microns is desirable in some aspects in order to maximize delivery to the lung while minimizing systemic delivery.
  • This property of the particles is measured according to methods known in the art. For example, it is measured as Mass Median Aerodynamic Diameter (MMAD).
  • MMAD Mass Median Aerodynamic Diameter
  • Micronized particles of this size can be produced by methods known in the art, for example by mechanical grinding (milling), spray-drying, freeze-drying, or lyophilization.
  • the average mean diameter of the particles can be from 0.5 to 10 microns, from 0.5 to 6 microns or from 5 to 10 microns. Regardless of whether the formulation is a liquid or dry powder, according to the methods of the disclosure it is dispensed in a manner suitable to produce particles or droplets of respirable size (e.g., 10 microns or less average mean diameter, preferably 5 to 10 microns where the lung itself is the therapeutic target, or less than 5 microns for deep lung delivery where systemic
  • the amount of drug released in fine, inhalable particles from a delivery device is referred to as the fine particle fraction (FPF) of the formulation.
  • the FPF is the percentage of the fine particle dose relative to the total amount of drug released.
  • Impaction occurs when the momentum of an inhaled particle is large enough that the particle does not follow the air stream and encounters a physiological surface.
  • sedimentation occurs primarily in the deep lung when very small particles which have traveled with the inhaled air stream encounter physiological surfaces as a result of random diffusion within the air stream.
  • a compound of the present disclosure can be deposited either by impaction (in the upper airways) or by sedimentation (in the alveoli).
  • the aerosol compositions of the disclosure are suitable for administration to a subject by conventional means.
  • the aqueous aerosol formulations, dry powder aerosol formulations, and propellant-based aerosol formulations provided by the instant disclosure, and described in further detail herein, comprise a compound of the present disclosure at a concentration of about 0.05 mg/ml to about 500 mg/ml, from about 0.05 mg/ml to about 250 mg/ml, or from about 0.05 mg/ml to about 100 mg/ml, or from about 0.01 to 5% or from about 0.01 to 2.5% based on total weight of the dry powder.
  • the aerosol formulation of the disclosure can be an aqueous formulation.
  • a compound of the present disclosure can be present as a suspension or emulsion in the aqueous formulation.
  • the aerosol composition of the disclosure can be in the form of a nebulizable aqueous, organic or aqueous/organic dispersion and the inhalation device is a nebulizer.
  • a number of nebulizers are available to aerosolize the formulations.
  • compressor-driven nebulizers use jet technology and use compressed air to generate the liquid aerosol.
  • Ultrasonic nebulizers rely on vibrations of a piezoelectric crystal to generate respirable liquid droplets and are commercially available from, for example, Omron Healthcare, Inc. and DeVilbiss Health Care, Inc. Nebulized solutions can be breathed directly from the nebulizing device or the nebulizing device can be attached to a face mask, tent or intermittent positive pressure breathing machine.
  • the nebulizer may be, for example, a conventional pneumatic nebulizer such as an airjet nebulizer, or an ultrasonic nebulizer, which may contain, for example, from 1 to 50 ml, commonly 1 to 10 ml, of the dispersion; or a hand-held nebulizer, for example an electronically controlled device such as an AERx (ex Aradigm, US) or a mechanical device such as a RESPIMAT (Boehringer Ingelheim) nebulizer which allows much smaller nebulized volumes, e.g.
  • a conventional pneumatic nebulizer such as an airjet nebulizer, or an ultrasonic nebulizer, which may contain, for example, from 1 to 50 ml, commonly 1 to 10 ml, of the dispersion
  • a hand-held nebulizer for example an electronically controlled device such as an AERx (ex Aradigm, US) or a mechanical device such as a
  • a pharmaceutical composition of the disclosure can be in the form of an aqueous aerosol formulation suitable for nasal delivery.
  • Formulations suitable for nasal inhalation are known in the art.
  • a nasal aerosol spray contains a compound a compound of the present disclosure, a water soluble diluent such as an organic acid, and a thickening agent such as a natural or synthetic polymer or an oil substance comprising the oil phase of an emulsion.
  • the compounds of the disclosure can also be administered in a vaporizer that delivers a volume of vapor containing a compound of the disclosure.
  • the vaporizer can be battery operated and designed to deliver an effective dosage of a compound of the present disclosure.
  • An aerosol formulation of the disclosure can be a dry powder formulation.
  • the dry powder formulation is suitable for use in either a dry powder inhaler device (DPI) or a pressurized metered dose inhaler (pMDI).
  • the active agent(s) can be present as a fine powder with a pharmaceutically acceptable carrier.
  • fine refers to a particle size in the inhalable range, as discussed herein.
  • a compound of the present disclosure can be micronized such that the particles have an average mean diameter in the range of 10 microns or less.
  • the particle size can be from 5 to 10 microns (average mean diameter) or more particularly from 3 to 5 microns in order to maximize delivery to the lung itself.
  • the particle size can be less than 5 microns, or preferably between 0.5 and 6 microns for deep lung delivery where systemic administration via the pulmonary route is desired.
  • Suitable carriers for use in the preparation of dry powders for inhalation are known in the art and include, for example saccharides, such as monosaccharides, disaccharides, polysaccharides and sugar alcohols such as arabinose, glucose, fructose, ribose, mannose, sucrose, trehalose, lactose, maltose, starches, dextran, and mannitol.
  • the carrier can be lactose, particularly in the form of the monohydrate.
  • the carrier particles can be of larger size (greater than 10 microns) so as to avoid deposition of the carrier material in the deep lung.
  • the average mean diameter particle size of the carrier material can be from 10 to 1000 microns or from 20 to 500 microns.
  • the carrier material can be a crystalline carrier material.
  • the crystalline carrier material can be one which is at least 95%, preferably 99% crystalline and in which no or substantially no water is absorbed by the carrier under conditions of 80% or lower relative humidity at room temperature. Examples of such crystalline carriers are lactose monohydrate and glucose monohydrate.
  • the amount of carrier is from 1 to 99% or more of the formulation by dry weight of the powder, preferably 5 to 99%, 10 to 99%, 20 to 99%, 30 to 99%, 40 to 99%, or 50 to 99%.
  • the dry powder formulation can further comprise one or more additives.
  • the one or more additives can be selected from the group consisting of l,2-dimyristoyl-sn-glycero-3- phosphocholine (DMPC), l,2-distearoyl-sn-glycero-3-phosphocholine (DSPC),
  • dipalmitoylphosphatidylcholine DPPC
  • dipalmitoylphosphatidylethanolamine DPPE
  • dipalmitoylphosphatidylethanolamine-PEG DPPE-PEG
  • dipalmitoylphosphatidylglycerol DPPG
  • l,2-dipalmitoyl-sn-glycero-3-phosphocholine DPPC
  • cholesterol lecithin
  • stearic acid and oleic acid
  • the additive can be human serum albumin.
  • the additive can be selected from the additives described herein.
  • the one or more additives can comprise or consist of magnesium stearate.
  • the magnesium stearate can be present in amounts of 0.001 to 10% by dry weight of the powder, preferably in amounts of from 0.01 to 5% or 0.01 to 2%.
  • the additive can comprise or consist of a phospholipid, such as lecithin (which is a mixture of phosphatidyl cholines) in an amount of 0.1% to 1% by dry weight of the powder, preferably 0.2% to 0.6%.
  • the additive can be coated onto the carrier material prior to or simultaneously with a step of blending the carrier with the particles of the compounds of the present disclosure.
  • Low energy devices for mixing dry powders to form blends include, for example, V-blenders, double cone blenders, slant cone blenders, cube blenders, bin blenders, horizontal or vertical drum blenders, static continuous blenders, and dynamic continuous blenders.
  • Other, higher energy devices include high shear mixers known to those skilled in the art.
  • the dry powder can be contained in capsules of gelatin or plastic, or in blisters, suitable for use in a DPI device, preferably in dosage units together with the carrier in amounts to bring the total weight of powder in each capsule to from 1 mg to 50 mg.
  • the dry powder may be contained in a reservoir of a multi-dose DPI device.
  • Preferred DPI devices include a blister based device such as the GyroHaler® or the OmniHaler® (both from Vectura), a reservoir based device such as the Clickhaler® or Duohaler® (Vectura), and the ARCUS® inhaler (Civitas Therapeutics).
  • the DPI technology can be selected from PulmatrixTM, and Hovone Twincaps and XCapsTM.
  • the DPI device can be selected from the group consisting of ArcusTM, AspirairTM, AxahalerTM, BreezhalerTM, ClickhalerTM, Conix DryTM, CricketTM, DreamboatTM, GenuairTM, GeminiTM, InspiromaticTM, iSPERSETM, MicroDoseTM, Next DPITM, ProhalerTM, PulmojetTM, PulvinalTM, SolisTM, TaifunTM, Taper DryTM, TrivaiTM, NovolizerTM, PodhalerTM, SkyehalerTM, SpiromaxTM, Twincaps/FlowcapsTM, and TurbuhalerTM.
  • the dry powder device can be adapted to deliver the dry powder from a capsule or blister containing a dosage unit of the dry powder or a multi-dose dry powder inhalation device adapted to deliver, for example, 5-25 mg of dry powder per actuation.
  • the particle size of the compound of the present disclosure can be reduced to the desired level by conventional methods, for example by grinding in an air-jet mill, ball mill or vibrator mill, microprecipitation, spray drying, lyophilization or recrystallization from supercritical media. Grinding in this context refers to micronization of drug particles by mechanical means. Micronization techniques do not require making a solution, slurry, or suspension of the drug. Instead, the drug particles are mechanically reduced in size. Due to the relatively high energy that is employed by micronization, in certain aspects it is desirable to include a carrier material in a co-micronized mixture with a compound of the present disclosure. In this context, the carrier material absorbs some of the energy of micronization which otherwise could adversely affect the structure of the active agent.
  • Spray drying generally involves making a solution, slurry, or suspension of a compound of the present disclosure, atomizing the solution, slurry, or suspension, to form particles and then drying the particles.
  • the powder formulation comprising a compound of the present disclosure can be made by spray drying an aqueous dispersion of the compound to form a dry powder consisting of aggregated particles of the compound having a size suitable for pulmonary delivery, as described herein.
  • the aggregate particle size can be adjusted (increased or decreased) to target either the deep lung or upper respiratory sites, such as the upper bronchial region or nasal mucosa. This can be accomplished, for example, by increasing the concentration of the compound of the present disclosure in the spray-dried dispersion or by increasing the droplet size generated by the spray dryer.
  • the dry powder can be made by freeze-drying the aqueous drug dispersion, or by a combination of spray-drying and freeze-drying.
  • the dry powder formulation can be made by freeze-drying an aqueous dispersion of a compound of the present disclosure, and one or more optional additives.
  • the powders can contain aggregates of a compound of the present disclosure and an additive, if present, wherein the aggregates are within a respirable size range as described herein.
  • the aqueous dispersion of a compound of the present disclosure and the one or more optional additives can further comprise a dissolved diluent such as lactose or mannitol such that when the dispersion is freeze-dried, respirable diluent particles, each containing at least one embedded drug particle and additive particle, if present, are formed.
  • the dry powder can comprise liposomes loaded with a compound of the present disclosure. Drug-loaded liposomes can be produced by methods known in the art, for example using the technique described for tacrolimus in M. Chougale, et al. Int. J.
  • HSPC hydrogenated phosphatidylcholine
  • cholesterol a compound of the present disclosure, hydrogenated phosphatidylcholine (HSPC), and cholesterol are dissolved in a mixture of methanol and chloroform and then subjected to dry thin film formation, e.g., in
  • the liposomes are hydrated and the liposomal dispersion is passed through a high-pressure homogenizer for size reduction.
  • the resultant pellets are characterized for vesicle size and percent drug entrapment and pellets equivalent to the desired amount of a compound of the present disclosure are then dispersed in a suitable medium and subjected to spray-drying to obtain particles of the desired size for inhalation.
  • the spray dried powder can be filled into capsules, canisters, or blister packs for administration.
  • the dry powder aerosol formulations of the disclosure can be delivered using any suitable dry powder inhaler device.
  • suitable dry powder inhaler device examples include the Aerolizer® Clickhaler®, Diskus®, Duohaler®, Flexhaler®, GyroHaler®, Handihaler®, Neohaler®, OmniHaler®, PressairTM, Rotahaler®, Turbuhaler® , and Twisthaler®.
  • the device can be selected from a blister based device such as the GyroHaler® or the OmniHaler® or a reservoir based device such as the Clickhaler® or Duohaler® all by Vectura.
  • the device can be a breath-actuated ARCUS® inhaler (Civitas Therapeutics).
  • the carrier if present, is selected from the group consisting of a saccharide and a sugar alcohol.
  • the carrier if present, can be lactose.
  • the compounds of the present disclosure can be formulated in a propellant-based formulation which may also be referred to generically herein as "a pMDI formulation".
  • the propellant-based formulation is prepared by methods known in the art, for example by wet milling a compound of the present disclosure, and an optional additive, in liquid propellant, either at ambient pressure or under high pressure conditions.
  • the additive can be a surfactant which serves to prevent aggregation (caking or crystallization), to facilitate uniform dosing, and (or alternatively) to provide a favorable fine particle fraction (FPF).
  • the surfactant can be selected from sorbitan trioleate, sorbitan monooleate, or oleic acid.
  • dry powders containing particles of compound of the present disclosure are prepared by spray-drying or freeze-drying aqueous dispersions of the particles of the present disclosure as described herein and the resultant powders dispersed into suitable propellants for use in conventional pressurized metered dose inhalers (pMDIs).
  • the compounds of the present disclosure can be in solution or in suspension in the propellant.
  • in suspension refers to where a compound of the present disclosure is present in particulate form dispersed in the propellant.
  • the compound of the present disclosure can be micronized and can be present in suspension in the propellant.
  • the formulation can further comprise a wetting agent or co-solvent such as ethanol.
  • the formulation can further comprise a polyhydroxy alcohol such as propylene glycol.
  • Suitable propellants include, for example, halogen- substituted hydrocarbons, for example fluorine-substituted methanes, ethanes, propanes, butanes, cyclopropanes or cyclobutanes, particularly 1, 1, 1,2-tetrafluoroethane (HFA134a) and 1, 1, 1,2,3, 3,3-heptafluoropropane (HFA227), or mixtures thereof.
  • halogen- substituted hydrocarbons for example fluorine-substituted methanes, ethanes, propanes, butanes, cyclopropanes or cyclobutanes, particularly 1, 1, 1,2-tetrafluoroethane (HFA134a) and 1, 1, 1,2,3, 3,3-heptafluoropropane (HFA227), or mixtures thereof.
  • the formulation can comprise micronized particles of a compound of the present disclosure, ethanol, a suitable propellant such as HFA 134a, HFA 227, or a mixture of suitable propellants, and optionally one or more surfactants.
  • a suitable propellant such as HFA 134a, HFA 227, or a mixture of suitable propellants, and optionally one or more surfactants.
  • the composition can be a propellant-based formulation suitable for delivery by a device such as a pressurized metered dose inhaler (pMDI).
  • the formulation can comprise a compound of the present disclosure, alone or together with a second active agent, a propellant, and a vegetable oil or pharmaceutically acceptable derivative of a vegetable oil.
  • the propellant is preferably selected from 1, 1, 1,2-tetrafluoroethane (HFA134a) and
  • the vegetable oil can be olive oil, safflower oil, and soybean oil.
  • the formulation can comprise micronized particles of a compound of the present disclosure, a micronized bulking agent, a suitable propellant such as HFA 134a, HFA 227, or a mixture of suitable propellants, and optionally one or more surfactants.
  • a suitable propellant such as HFA 134a, HFA 227, or a mixture of suitable propellants, and optionally one or more surfactants.
  • the formulation can further comprise a lubricant.
  • the formulation can comprise a compound of the present disclosure, a propellant, and a vegetable oil.
  • the formulation does not comprise an additive or surfactant.
  • the formulation does not comprise ethanol, a polyhydroxy alcohol (e.g., propylene glycol), or a surfactant (e.g., sorbitan trioleate, sorbitan monooleate, or oleic acid).
  • a pharmaceutical composition of the disclosure can be in in the form of an orally acceptable dosage form including, but not limited to, capsules, tablets, buccal forms, troches, lozenges, and oral liquids in the form of emulsions, aqueous suspensions, dispersions or solutions.
  • Capsules may contain mixtures of a compound of the present disclosure with inert fillers and/or diluents such as the pharmaceutically acceptable starches (e.g., corn, potato or tapioca starch), sugars, artificial sweetening agents, powdered celluloses, such as crystalline and micro-crystalline celluloses, flours, gelatins, gums, etc.
  • carriers which are commonly used include lactose and corn starch.
  • Lubricating agents such as magnesium stearate, can also be added.
  • useful diluents include lactose and dried corn starch.
  • the compound of the present disclosure may be suspended or dissolved in an oily phase is combined with emulsifying and/or suspending agents. If desired, certain sweetening and/or flavoring and/or coloring agents may be added.
  • a pharmaceutical composition of the disclosure can be in the form of a tablet.
  • the tablet can comprise a unit dosage of a compound of the present disclosure together with an inert diluent or carrier such as a sugar or sugar alcohol, for example lactose, sucrose, sorbitol or mannitol.
  • the tablet can further comprise a non-sugar derived diluent such as sodium carbonate, calcium phosphate, calcium carbonate, or a cellulose or derivative thereof such as methyl cellulose, ethyl cellulose, hydroxypropyl methyl cellulose, and starches such as corn starch.
  • the tablet can further comprise binding and granulating agents such as
  • polyvinylpyrrolidone e.g. swellable crosslinked polymers such as crosslinked carboxymethylcellulose
  • disintegrants e.g. swellable crosslinked polymers such as crosslinked carboxymethylcellulose
  • lubricating agents e.g. stearates
  • preservatives e.g. parabens
  • antioxidants e.g. BHT
  • buffering agents for example phosphate or citrate buffers
  • effervescent agents such as citrate/bicarbonate mixtures.
  • the tablet can be a coated tablet.
  • the coating can be a protective film coating (e.g. a wax or varnish) or a coating designed to control the release of the active agent, for example a delayed release (release of the active after a predetermined lag time following ingestion) or release at a particular location in the gastrointestinal tract.
  • the latter can be achieved, for example, using enteric film coatings such as those sold under the brand name Eudragit®.
  • Tablet formulations may be made by conventional compression, wet granulation or dry granulation methods and utilize pharmaceutically acceptable diluents, binding agents, lubricants, disintegrants, surface modifying agents (including surfactants), suspending or stabilizing agents, including, but not limited to, magnesium stearate, stearic acid, talc, sodium lauryl sulfate, microcrystalline cellulose, carboxymethylcellulose calcium,
  • polyvinylpyrrolidone gelatin, alginic acid, acacia gum, xanthan gum, sodium citrate, complex silicates, calcium carbonate, glycine, dextrin, sucrose, sorbitol, dicalcium phosphate, calcium sulfate, lactose, kaolin, mannitol, sodium chloride, talc, dry starches and powdered sugar.
  • Preferred surface modifying agents include nonionic and anionic surface modifying agents.
  • surface modifying agents include, but are not limited to, poloxamer 188, benzalkonium chloride, calcium stearate, cetostearyl alcohol, cetomacrogol emulsifying wax, sorbitan esters, colloidal silicon dioxide, phosphates, sodium
  • dodecylsulfate magnesium aluminum silicate, and triethanolamine.
  • a pharmaceutical composition of the disclosure can be in the form of a hard or soft gelatin capsule.
  • the compound of the present disclosure may be in a solid, semi-solid, or liquid form.
  • a pharmaceutical composition of the disclosure can be in the form of a sterile aqueous solution or dispersion suitable for parenteral administration.
  • parenteral as used herein includes subcutaneous, intracutaneous, intravenous, intramuscular, intra-articular, intraarterial, intrasynovial, intrasternal, intrathecal, intralesional and intracranial injection or infusion techniques.
  • a pharmaceutical composition of the disclosure can be in the form of a sterile aqueous solution or dispersion suitable for administration by either direct injection or by addition to sterile infusion fluids for intravenous infusion, and comprises a solvent or dispersion medium containing, water, ethanol, a polyol (e.g., glycerol, propylene glycol and liquid polyethylene glycol), suitable mixtures thereof, or one or more vegetable oils.
  • a solvent or dispersion medium containing, water, ethanol, a polyol (e.g., glycerol, propylene glycol and liquid polyethylene glycol), suitable mixtures thereof, or one or more vegetable oils.
  • Solutions or suspensions of the compound of the present disclosure as a free base or pharmacologically acceptable salt can be prepared in water suitably mixed with a surfactant.
  • a surfactant examples include sodium sulfate, sodium sulfate, sodium sulfate, sodium sulfate, sodium sulfate, sodium sulfate, sodium sulfate, sodium sulfate, sodium sulfate, sodium bicarbonate, sodium sulfate, sodium sulfate, sodium sulfate, sodium sulfate, sodium bicarbonate, sodium bicarbonate, sodium bicarbonate, sodium sulfate, sodium sulfate, sodium sulfate, sodium sulfate, sodium sulfate, sodium sulfate, sodium sulfate, sodium sulfate, sodium sulfate, sodium sulfate, sodium sulfate, sodium sulfate, sodium sulfate, sodium sul
  • a pharmaceutical composition of the disclosure can be in the form of a lotion, ointment, cream, salve, gel, foam, patch, suspension, solution, or suppository (rectal or vaginal) for transdermal or transmucosal administration and can include a carrier that is inert and non-toxic to the skin.
  • Transdermal and transmucosal administration includes administration across the surface of the body and the inner linings of bodily passages such as epithelial and mucosal tissues.
  • the carrier can be in the form of a cream or ointment, a paste, or a gel.
  • the creams and ointments can be viscous liquid or semisolid emulsions of either the oil-in-water or water-in-oil type.
  • the pastes may be comprised of absorptive powders dispersed in petroleum or hydrophilic petroleum.
  • Suppository formulations may be made from traditional materials, including cocoa butter, with or without the addition of waxes to alter the suppository's melting point, and glycerin.
  • Water soluble suppository bases such as polyethylene glycols of various molecular weights, may be included in the formulation.
  • the transmucosal or transdermal formulations can further comprise one or more penetrants appropriate to the barrier to be permeated.
  • the pharmaceutical compositions of the present disclosure can further comprise one or more additives in addition to any carrier or diluent (such as lactose or mannitol) that is present in the formulation.
  • the one or more additives can comprise or consist of one or more surfactants.
  • Surfactants typically have one or more long aliphatic chains such as fatty acids which enables them to insert directly into the lipid structures of cells to enhance drug penetration and absorption.
  • An empirical parameter commonly used to characterize the relative hydrophilicity and hydrophobicity of surfactants is the hydrophilic-lipophilic balance ("HLB" value).
  • HLB values Surfactants with lower HLB values are more hydrophobic, and have greater solubility in oils, while surfactants with higher HLB values are more hydrophilic, and have greater solubility in aqueous solutions.
  • hydrophilic surfactants are generally considered to be those compounds having an HLB value greater than about 10
  • hydrophobic surfactants are generally those having an HLB value less than about 10.
  • these HLB values are merely a guide since for many surfactants, the HLB values can differ by as much as about 8 HLB units, depending upon the empirical method chosen to determine the HLB value.
  • surfactants for use in the compositions of the disclosure are polyethylene glycol (PEG)-fatty acids and PEG- fatty acid mono and diesters, PEG glycerol esters, alcohol-oil transesterification products, polyglyceryl fatty acids, propylene glycol fatty acid esters, sterol and sterol derivatives, polyethylene glycol sorbitan fatty acid esters, polyethylene glycol alkyl ethers, sugar and its derivatives, polyethylene glycol alkyl phenols, polyoxyethylene-polyoxypropylene (POE-POP) block copolymers, sorbitan fatty acid esters, ionic surfactants, fat-soluble vitamins and their salts, water-soluble vitamins and their amphiphilic derivatives, amino acids and their salts, and organic acids and their esters and anhydrides.
  • PEG polyethylene glycol
  • PEG-fatty acid mono and diesters PEG glycerol esters
  • alcohol-oil transesterification products polyglyce
  • the present disclosure also provides packaging and kits comprising pharmaceutical compositions of the present disclosure.
  • the kit can comprise one or more containers selected from the group consisting of a bottle, a vial, an ampoule, a blister pack, and a syringe.
  • the kit can further include one or more of instructions for use in treating and/or preventing a disease, condition or disorder of the present disclosure, one or more syringes, one or more applicators, or a sterile solution suitable for reconstituting a pharmaceutical composition of the present disclosure.
  • Example 1 HTS assay of MEF-EV Cell.
  • JTC-801 was identified in a high throughput cell viability screen using TSC2-/- mouse embryonic fibroblasts (MEF-EV) cells.
  • MEF cells derived from TSC2 -/- knockout mouse embryos were infected with a retrovirus vector encoding the hygromycin antibiotic resistance gene (MEF-EV) or the same retrovirus vector also encoding TSC2 (MEF-TSC2).
  • MEF-EV hygromycin antibiotic resistance gene
  • MEF-TSC2 retrovirus vector also encoding TSC2
  • the MEF-EV and MEF-TSC2 line were then established by hygromycin selection.
  • Cells were expanded in DMEM containing 10% FBS (Omega Scientific) and 2mM L-Glutamine. Frozen stocks of cells were prepared for direct use in the HTS assay.
  • Test compounds (5 ⁇ stock solution, 6 x desired final well concentration) were dispensed to 384-well assay plates (Corning 3712) using a Biomek FX liquid handler. MEF- EV cells (1000 cells per well in 25 ⁇ ⁇ of media) were added to these pre-formatted plates using a Thermo Wellmate non-contact dispensing system with a standard bore cassette head. Plates were incubated for 72h at 37°C under an atmosphere of 5% CO2 in a humidified incubator.
  • Example 2 HTS concentration response curves in MEF and AML cells.
  • a second HTS screen was performed using additional cell lines: MEF EV, MEF TSC2, AML-102 (TSC2 deficient) and AML-103 (TSC2 restored).
  • MEF EV MEF EV
  • MEF TSC2 MEF TSC2
  • AML-102 TSC2 deficient
  • AML-103 TSC2 restored
  • Test compounds (5 ⁇ stock solution, 6 x desired final well concentration) were diluted to achieve a 10 point concentration response curve (40000-78nM; 3-fold dilution series). Plates were incubated for 72h at 37°C under an atmosphere of 5% CO2 in a humidified incubator.
  • JTC-801 displays anti-proliferative in cell lines.
  • JTC- 801 is more effective in cell lines hyper active for mTOR (i.e., lacking TSC2 compared with TSC2 rescue cell lines).
  • Example 3 Concentration response curves JTC-801 in a panel of cancer cell lines.
  • a panel of cancer cell lines (Mia-PaCa-2, H4, 22RV 1 , HCT 1 16, DU 145, MCF-7 and HI 299) representing different tumor types (pancreatic, neuroglioma, colon, prostate, breast and lung) was used to test JTC-801 single agent activity.
  • JTC-801 50 ⁇ stock solution, 4 x desired final well concentration
  • a 10-point concentration response curve 10000-19.5nM; 2-fold dilution series
  • Plates were incubated for 72h at 37°C under an atmosphere of 5% CO2 in a humidified incubator.
  • Cell viability was determined with CellTiter-Glo® luminescence assay (Promega) as per the manufacturer's instructions. Viability was expressed as a percentage of DMSO only control cells (see FIG. 2).
  • Example 4 - HTS identifies combinatorial activity between pazopanib and JTC-801.
  • MEF EV cells were used to conduct drug combination studies to identify synergistic drug pairs. For screening, MEF EV cells were grown and prepared as described above. A panel of unapproved compounds were screened at 50 nM, 500 nM, 5000 nM (final concentration) in the presence or absence of pazopanib (5000 nM). Plates were incubated for 72h at 37°C under an atmosphere of 5% CO2 in a humidified incubator. Cell viability was determined with CellTiter-Glo® luminescence assay (Promega) as per the manufacturer's instructions. Viability was expressed as a percentage of untreated control cells. The viability of the unapproved drug alone was compared to the unapproved drug + pazopanib.
  • JTC-801 was identified as significantly reducing MEF EV cell viability only in the presence of pazopanib (see FIG. 3).
  • Example 5 Synergistic activity of JTC-801 in TSC2 deficient cell lines.
  • JTC-801 Combinations of JTC-801 along with other compounds was performed.
  • MEF EV, MEF TSC2, ELT-3 EV, ELT-3 TSC2, AML- 102 and AML-103 cell lines were grown and used for screening as described above.
  • JTC-801 50 ⁇ stock solution, 8 x desired final well concentration
  • JTC-801 was diluted to achieve an 8-point concentration response curve (5000-2.3nM; 3-fold dilution series).
  • Cells treated at each concentration of JTC-801 were additionally co- treated with DMSO or with a second compound (5000-2.3nM; 3-fold dilution series). This generated an 8x8 drug concentration matrix, with each concentration of JTC-801 tested with each of the 8 concentrations of the other compound.
  • Example 6 Combinatorial activity of JTC-801 and pazopanib in cancer cell lines.
  • a panel of cancer cell lines (Mia-PaCa-2, H4, 22RV1, HCT1 16, DU145, MCF-7 and HI 299) representing different tumor types (pancreatic, neuroglioma, colon, prostate, breast and lung) were used to test JTC-801 combinatorial activity.
  • Cell lines were seeded into 96 well plates as described above. Cancer cells were treated with JTC-801. JTC-801 (50 ⁇ stock solution, 4 x desired final well concentration) and diluted to achieve a 10-point concentration response curve ( 10000-19.5nM; 2-fold dilution series) in the presence or absence of pazopanib (fixed concentration of 5000 nM). Plates were incubated for 72h at 37°C under an atmosphere of 5% CO2 in a humidified incubator. Cell viability was determined with CellTiter-Glo® luminescence assay (Promega) as per the manufacturer's instructions.
  • Viability was expressed as a percentage of DMSO only control cells (see FIG. 5).
  • Example 7 - HTS identifies combinatorial activity between disulfiram and JTC-801
  • MEF EV cells were grown and prepared as described above.
  • a panel of test compounds was screened at 50 nM, 500 nM, 5000 nM (final concentration) in the presence or absence of disulfiram (100 nM). Plates were incubated for 72h at 37°C under an atmosphere of 5% CO2 in a humidified incubator. Cell viability was determined with CellTiter-Glo® luminescence assay (Promega) as per the manufacturer's instructions. Viability was expressed as a percentage of untreated control cells. The viability of each test compound alone was compared to the test compound + disulfiram.
  • JTC-801 was identified as significantly reducing MEF EV cell viability only in the presence of disulfiram (see FIG. 6). These findings suggest that the combination of JTC-801 and disulfiram may be effective in suppressing proliferative diseases. Thus, this combination may be especially effective where disulfiram is efficacious as a single agent such as against pancreatic cancer (Kim et al. 2013, or breast cancer (Wiggins et al., 2015) or is being tested clinically, such as against metastatic non-small cell lung cancer (Nechushtan et al., 2015).
  • Example 8 - HTS identifies combinatorial activity between fulvestrant (Faslodex®) and JTC-801
  • MEF EV cells were grown and prepared as described above.
  • a panel of test compounds was screened at 50 nM, 500 nM, 5000 nM (final concentration) in the presence or absence of Faslodex (fulvestrant here-in) (5000 nM).
  • JTC-801 was identified as significantly reducing MEF EV cell viability only in the presence of fulvestrant (see FIG. 7).
  • JTC-801 Combinations of JTC-801 along with imatinib was performed in ELT-3 EV cells. Cells were grown and used for screening as described above. JTC-801 (50 ⁇ stock solution, 8 x desired final well concentration) was diluted to achieve an 8-point concentration response curve (5000-2.3 nM; 3-fold dilution series). Cells treated at each concentration of JTC-801 were additionally co-treated with DMSO or with a second compound (5000-2.3 nM; 3-fold dilution series). This generated an 8x8 drug concentration matrix, with each concentration of JTC-801 tested with each of the 8 concentrations of the other compound. Plates were incubated for 72h at 37°C under an atmosphere of 5% CO2 in a humidified incubator. Cell viability was determined with CellTiter-Glo® luminescence assay (Promega) as per the manufacturer's instructions. Viability was expressed as a percentage of DMSO only control cells (see FIG. 8).
  • JTC-801 and imatinib may be effective in suppressing proliferative diseases.
  • this combination may be especially effective where imatinib is used clinically, such as for chronic myelogenous leukemia, gastrointestinal stromal tumors and acute lymphoblastic leukemia, amongst others.
  • Example 10 Computational approaches identifies cancer tissue types predicted to be sensitive to JTC-801
  • the cancer cell lines signatures were compared to the JTC-801 gene expression profiles by using a ranked-based algorithm (Lamb et al. 2006). Briefly, each cancer cell signature was queried against the rank-ordered drug expression profile to quantitatively measure the similarity between both profiles, and an enrichment score was calculated separately for the up-regulated and down-regulated gene sets. This analysis proposes that if the up-regulated cancer genes match the bottom (down-regulated) of the drug expression profile and the down-regulated cancer genes match the top (up-regulated) of the drug expression profile, it is considered that the drug-cancer pair has complementary expression profiles.
  • FIGs. 9A-9B shows the output of the analysis for JTC-801.
  • JTC-801 is considered to be a potential treatment for the cancers shown in FIGs. 9A-9B.
  • JTC-801 was modified with a linker to afford linker compounds JTC-801-1, JTC-801 - 2, JTC-801 -3, and JTC-801 -4 (FIG. 10). Cell viability in the presence of these compounds was determined with CellTiter-Glo® luminescence assay (Promega) as per the
  • One capture compound was selected to identify specific protein binders in the CCMS experiments using JTC-801 as a competitor ligand. For the studies with cell homogenate the supernatant and protein pellet were isolated into individual fractions and evaluated separately. Proteins that are detected by LC-S in the capture assay and that are significantly diminished in competition control experiments are considered to be specific binders. These specific binders were further subjected to stringent data analysis criteria to determine specificity after unbiased data evaluation. Specific protein binders were ranked according to their fold change (FC) values in the capture experiments.
  • FC fold change

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

La présente invention concerne des compositions de 4-aminoquinoline (un exemple de composé étant JTC-801) ayant une activité cytotoxique et/ou anti-proliférative, et des méthodes d'utilisation des compositions pour le traitement et/ou la prévention de maladies, d'affections ou de pathologies prolifératives chez un sujet ayant besoin d'un tel traitement. Les maladies peuvent être le cancer, les maladies neurodégénératives, ou les maladies caractérisées par une anomalie de la voie de signalisation de mTOR. L'invention concerne également des méthodes d'inhibition de l'activité ACOX3 (acyl-coenzyme A oxydase des peroxysomes).
PCT/US2015/038287 2014-07-02 2015-06-29 Compositions de 4-aminoquinoline et méthodes d'utilisation WO2016003886A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201462020167P 2014-07-02 2014-07-02
US62/020,167 2014-07-02

Publications (1)

Publication Number Publication Date
WO2016003886A1 true WO2016003886A1 (fr) 2016-01-07

Family

ID=55019874

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2015/038287 WO2016003886A1 (fr) 2014-07-02 2015-06-29 Compositions de 4-aminoquinoline et méthodes d'utilisation

Country Status (1)

Country Link
WO (1) WO2016003886A1 (fr)

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005004896A1 (fr) * 2003-07-01 2005-01-20 Ufpeptides S.R.L. Antagonistes du recepteur nop pour le traitement de la maladie de parkinson
US20060030565A1 (en) * 1998-03-26 2006-02-09 Japan Tobacco Inc. Amide derivatives and nociceptin antagonists
US20100272717A1 (en) * 2007-12-13 2010-10-28 Novartis Ag Combinations of therapeutic agents for treating cancer

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060030565A1 (en) * 1998-03-26 2006-02-09 Japan Tobacco Inc. Amide derivatives and nociceptin antagonists
WO2005004896A1 (fr) * 2003-07-01 2005-01-20 Ufpeptides S.R.L. Antagonistes du recepteur nop pour le traitement de la maladie de parkinson
US20100272717A1 (en) * 2007-12-13 2010-10-28 Novartis Ag Combinations of therapeutic agents for treating cancer

Similar Documents

Publication Publication Date Title
JP7019422B2 (ja) 癌を治療するための方法
US11266654B2 (en) Apilimod compositions and methods for using same
US11166944B2 (en) Apilimod compositions and methods for using same in the treatment of renal cancer
JP2022088616A (ja) 再発性神経膠腫および進行性の二次性脳腫瘍からなる群から選択される悪性腫瘍の治療に使用するための薬剤の調製方法
TWI746449B (zh) 使用阿吡莫德治療癌症之方法
US20180078561A1 (en) Active metabolites of apilimod and uses thereof
CN107427522B (zh) 用于治疗黑素瘤的阿吡莫德
WO2016003886A1 (fr) Compositions de 4-aminoquinoline et méthodes d&#39;utilisation
NZ722491B2 (en) Apilimod compositions and methods for using same

Legal Events

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

Ref document number: 15815836

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 15815836

Country of ref document: EP

Kind code of ref document: A1