WO2016025651A1 - Combinations of an erk inhibitor and a tor inhibitor and related methods - Google Patents

Combinations of an erk inhibitor and a tor inhibitor and related methods Download PDF

Info

Publication number
WO2016025651A1
WO2016025651A1 PCT/US2015/044931 US2015044931W WO2016025651A1 WO 2016025651 A1 WO2016025651 A1 WO 2016025651A1 US 2015044931 W US2015044931 W US 2015044931W WO 2016025651 A1 WO2016025651 A1 WO 2016025651A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
cancer
substituted
unsubstituted
pharmaceutically acceptable
Prior art date
Application number
PCT/US2015/044931
Other languages
French (fr)
Inventor
Ida ARONCHIK
Carmen Maria BARNES
Gordon L. BRAY
Ellen H. Filvaroff
Original Assignee
Celgene Avilomics Research, 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 Celgene Avilomics Research, Inc. filed Critical Celgene Avilomics Research, Inc.
Publication of WO2016025651A1 publication Critical patent/WO2016025651A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/365Lactones
    • A61K31/366Lactones having six-membered rings, e.g. delta-lactones
    • 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
    • A61K31/4353Heterocyclic 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 ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/436Heterocyclic 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 ortho- or peri-condensed with heterocyclic ring systems the heterocyclic ring system containing a six-membered ring having oxygen as a ring hetero atom, e.g. rapamycin
    • 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
    • A61K31/4985Pyrazines or piperazines ortho- or peri-condensed with heterocyclic ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca

Definitions

  • the present invention provides methods of treating, stabilizing or lessening the severity or progression of a disease or disorder associated with one or both of ERK1 and ERK2 protein kinase.
  • Protein kinases constitute a large family of structurally related enzymes that are responsible for the control of a variety of signal transduction processes within the cell. Protein kinases are thought to have evolved from a common ancestral gene due to the conservation of their structure and catalytic function. Almost all kinases contain a similar 250-300 amino acid catalytic domain. The kinases may be categorized into families by the substrates they phosphorylate (e.g., protein-tyrosine, protein-serine/threonine, lipids, etc.).
  • the processes involved in tumor growth, progression, and metastasis are mediated by signaling pathways that are activated in cancer cells.
  • the MAPK or Raf-Mek-ERK pathway plays a central role in regulating mammalian cell growth by relaying extracellular signals from ligand-bound cell surface tyrosine kinase receptors such as erbB family, PDGF, FGF, and VEGF receptor tyrosine kinase.
  • ligand-bound cell surface tyrosine kinase receptors such as erbB family, PDGF, FGF, and VEGF receptor tyrosine kinase.
  • Activation of the ERK occurs via a cascade of phosphorylation events that begins with activation of Ras.
  • Activation of Ras leads to the recruitment and activation of Raf, a serine-threonine kinase.
  • Raf Activated Raf then phosphorylates and activates MEK1/2, which then phosphorylates and activates one or both of ERK1 and ERK2.
  • MEK1/2 When activated, one or both of ERK1 and ERK2 phosphorylates several downstream targets involved in a multitude of cellular events including cytoskeletal changes and transcriptional activation.
  • the ERK/MAPK pathway is one of the most important for cell proliferation, and human tumor data suggest that the ERK/MAPK pathway is frequently activated in many tumors.
  • Ras genes which are upstream of one or both of ERK1 and ERK2, are mutated in several cancers including colorectal, melanoma, breast, lung, and pancreatic tumors.
  • High Ras activity is accompanied by elevated ERK activity in many human tumors.
  • activating mutations of BRAF a serine- threonine kinase of the Raf family, are associated with increased RAF, MEK, and ERK kinase activity.
  • Tumors types with the most frequent mutations in BRAF include melanomas (60%), thyroid cancers (greater than 40%) and colorectal cancers.
  • the present invention provides methods of treating, stabilizing or lessening the severity or progression of one or more diseases or disorders associated with one or both of ERK1 and ERK2 comprising administering to a patient in need thereof an inhibitor of one or both of ERK 1 and ERK2 in combination with a TOR (i.e., target of rapamycin) inhibitor.
  • the inhibitor of one or both of ERK1 and ERK2 is Compound 1 (N-(2-((2-(2- methoxy-5-methylpyridin-4-yl)amino)-5-(trifluoromethyl)pyrimidin-4-yl)amino)-5- methylphenyl)acrylamide):
  • Compound 1 N-(2-((2-((2-methoxy-5-methylpyridin-4-yl)amino)-5- (trifluoromethyl)pyrimidin-4-yl)amino)-5-methylphenyl)acrylamide, is designated as compound number I-90 in PCT patent application serial number PCT/US14/15256, filed February 7, 2014 and published as WO2014/124230 on August 14, 2014 (referred to herein as“the ‘230 publication,”) the entirety of which is hereby incorporated by reference. The synthesis of Compound 1 is described in detail at Example 94 of the‘230 publication.
  • Compound 1 is active in a variety of assays and therapeutic models demonstrating covalent, irreversible inhibition of one or both of ERK1 and ERK2 kinases (see, e.g., Table A of the ‘230 publication). Accordingly, Compound 1, or a pharmaceutically acceptable salt thereof, is useful for treating one or more disorders associated with activity of one or both of ERK1 and ERK2, as described in detail herein, infra.
  • FIGs 1A, 1B and 1C Panc1 (a.k.a., PANC-1) Pancreatic Cancer Cell Line: Panc 1 cells were treated with a 9-point dose dilution of Compound 1 or Compound 2 or both, ranging from 1.52 nM to 10000.00 nM and DMSO vehicle control.
  • Fig. 1A dose response to Compound 2 (trend line with triangles);
  • Fig. 1B dose response to Compound 1 (trend line with circles);
  • Fig. 1C combination treatment with Compound 1 and Compound 2 (trend line with circles).
  • FIG. 2A dose response to Compound 2
  • Fig. 2B dose response to Compound 1 (trend line with circles)
  • Fig. 2C combination treatment with Compound 1 and Compound 2 (trend line with circles).
  • FIGs 3A, 3B and 3C HS294T were treated with a 9-point dose dilution of Compound 1, Compound 2, or both, ranging from 1.52 nM to 10000.00 nM and DMSO vehicle control.
  • Fig. 3A dose response to Compound 2 (trend line with triangles);
  • Fig. 3B dose response to Compound 1 (trend line with circles);
  • Fig. 3C combination treatment with Compound 1 and Compound 2 (trend line with circles).
  • FIGs 4A, 4B and 4C HCT-116 cells were treated with a 9-point dose dilution of Compound 1 (Fig. 4A) or Compound 2 (Fig. 4B) ranging from 1.52 nM to 10000.00 nM and DMSO vehicle control. Combination treatment with Compound 1 and Compound 2 (Fig. 4C).
  • FIGs 5A, 5B and 5C NCI-H460 cells were treated with a 9-point dose dilution of Compound 1 (Fig. 5A) or Compound 2 (Fig. 5B) ranging from 1.52 nM to 10000.00 nM and DMSO vehicle control. Combination treatment with Compound 1 and Compound 2 (Fig. 5C).
  • FIGS. 6A, 6B and 6C NCI-H522 cells were treated with a 9-point dose dilution of Compound 1 (Fig. 6A) or Compound 2 (Fig. 6B) ranging from 1.52 nM to 10000.00 nM and DMSO vehicle control. Combination treatment with Compound 1 and Compound 2 (Fig. 6C).
  • FIGs 7A, 7B and 7C NCI-H1755 cells were treated with a 9-point dose dilution of Compound 1 (Fig. 7A) or Compound 2 (Fig. 7B) ranging from 1.52 nM to 10000.00 nM and DMSO vehicle control. Combination treatment with Compound 1 and Compound 2 (Fig. 7C).
  • FIG. 8 Emax values are shown against a panel of cancer cell lines. A moderate enhancement in Emax is seen when combining Compound 1 with Compound 2, a TOR inhibitor. Compound 1 does not kill any of the tested lines at 1 ⁇ M, and in combination with Compound 2, cell death is observed in 1/6 tested lines (i.e., NCI-H727). Emax in this line reached -16% when Compound 1 was combined with 1 ⁇ M Compound 2
  • Figure 9 The results of a Calcusyn analysis to detect synergy of Compound 1 and Compound 2 combination treatement are shown for a panel of B-raf mutant vemurafenib resistant melanoma cell lines.
  • FIG. 10 Activity in a BRAF-mutant vermurafenib-resistant melanoma cell line (i.e., cell line 8) colony forming assay is shown for the combination of Compound 1 and Compound 2.
  • FIG. 11 Activity in a BRAF-mutant vemurafenib-resistant melanoma cell line (i.e., cell line 3) colony forming assay is shown for the combination of Compound 1 and Compound 2.
  • Figure 12. CI values are shown for the combination of Compound 1 and Compound 2 in NRAS mutant melanoma cell lines A-D.
  • Figure 13. Depicts the results for Compound 1 (phosphate salt) and Compound 6 singly and in combination in the KRAS G12D cell line CRC PDX model.
  • FIG. 14 Volcano plot at section (a) for HCT-116 colorectal cancer cells treated with a combination of Compound 1 (9-point dose dilution) and Compound 2 at concentrations of 0.1 ⁇ M, 0.3 ⁇ M and 1.0 ⁇ M . Relevant dose response assay data are provided at section (b).
  • FIG. 15 Volcano plot at section (a) for HCT-116 colorectal cancer cells treated with a combination of Compound 1 (9-point dose dilution) and Compound 2 (at concentrations of 0.1 ⁇ M, 0.3 ⁇ M and 1.0 ⁇ M). Relevant dose response assay data are provided at section (b).
  • FIG. 16 Volcano plot at section (a) for HCT-116 colorectal cancer cells treated with a combination of Compound 1 (9-point dose dilution) and Compound 2 (at concentrations of 0.1 ⁇ M, 0.3 ⁇ M and 1.0 ⁇ M). Relevant dose response assay data are provided at section (b).
  • FIG. Volcano plot at section (a) for Mia Paca pancreatic cancer cells treated with a combination of Compound 1 (9-point dose dilution) and Compound 2 (at concentrations of 0.1 ⁇ M, 0.3 ⁇ M and 1.0 ⁇ M). Relevant dose response assay data are provided at section (b).
  • FIG. 1 Dose response assay data for Mia Paca pancreatic cancer cells treated with a combination of Compound 1 and Compound 2 (at concentrations of 0.1 ⁇ M, 0.3 ⁇ M and 1.0 ⁇ M).
  • FIG. 19 Volcano plot at section (a) for Mia Paca pancreatic cancer cells treated with a combination of Compound 1 (9-point dose dilution) and Compound 2 (at concentrations of 0.03 ⁇ M, 0.3 ⁇ M and 1.0 ⁇ M) (run 1). Relevant dose response assay data are provided at section (b).
  • FIG. 20 Volcano plot at section (a) for Mia Paca pancreatic cancer cells treated with a combination of Compound 1 (9-point dose dilution) and Compound 2 (at concentrations of 0.03 ⁇ M, 0.3 ⁇ M and 1.0 ⁇ M) (run 2). Relevant dose response assay data are provided at section (b).
  • FIG. 21 Mia PaCa Pancreatic Cancer Cell Line: Mia PaCa cells were treated with a 9-point dose dilution of Compound 1, or Compound 2, or both, ranging from 1.52 nM to 10000.00 nM, and DMSO vehicle control.
  • Fig. 21A dose response to Compound 1 alone (trend line with circles) or Compound 2 alone (trend line with triangles);
  • Fig. 21B combination treatment with Compound 1 and Compound 2 (trend line with circles) and untreated (trendline with triangles).
  • FIG. 22 Mia PaCa Pancreatic Cancer Cell Line: Mia PaCa cells were treated with a 9-point dose dilution of Compound 1, or Compound 2, or both, ranging from 1.52 nM to 10000.00 nM, and DMSO vehicle control.
  • Fig. 22A dose response to Compound 1 alone (trend line with circles) or Compound 2 alone (trend line with triangles);
  • Fig. 22B combination treatment with Compound 1 and Compound 2 (trend line with circles).
  • FIG. 23 HS294T Melanoma Cancer Cell Line: HS294T cells were treated with a 9-point dose dilution of Compound 1, or Compound 2, or both, ranging from 1.52 nM to 10000.00 nM, and DMSO vehicle control.
  • Fig. 23A dose response to Compound 1 alone (trend line with circles) or Compound 2 alone (trend line with triangles);
  • Fig. 23B combination treatment with Compound 1 and Compound 2 (trend line with circles).
  • FIG. 24 Volcano plot for Calu-1 lung cancer cells treated with a combination of Compound 1 (9-point dose dilution) and Compound 2 (at concentrations of 0.1 ⁇ M, 0.3 ⁇ M and 1.0 ⁇ M) (run 2).
  • FIGS 25A and 25B Volcano plot for HCT-116 colorectal cancer cells treated with a combination of Compound 1 (9-point dose dilution) and Compound 2 (at concentrations of 0.1 ⁇ M, 0.3 ⁇ M and 1.0 ⁇ M).
  • FIGS 26Aand 26B Volcano plot for Mia PaCa pancreatic cancer cells treated with a combination of Compound 1 (9-point dose dilution) and Compound 2 (at concentrations of 0.1 ⁇ M, 0.3 ⁇ M and 1.0 ⁇ M).
  • FIGS 27A, 27B, 27C and 27D Volcano plot for Calu-6 lung cancer cells treated with a combination of Compound 1 (9-point dose dilution) and Compound 2 (at concentrations of 0.1 ⁇ M, 0.3 ⁇ M and 1.0 ⁇ M).
  • FIGS 28A and 28B Volcano plot for NCI-H460 cells treated with a combination of Compound 1 (9-point dose dilution) and Compound 2 (at concentrations of 0.1 ⁇ M, 0.3 ⁇ M and 1.0 ⁇ M).
  • FIGS 29A and 29B Volcano plot for NCI-H727 cells treated with a combination of Compound 1 (9-point dose dilution) and Compound 2 (at concentrations of 0.1 ⁇ M, 0.3 ⁇ M and 1.0 ⁇ M).
  • the present invention provides methods of treating, stabilizing or lessening the severity or progression of one or more diseases or disorders associated with one or both of ERK1 and ERK2 comprising administering to a patient in need thereof an inhibitor of one or both of ERK1 and ERK2 in combination with a TOR inhibitor.
  • an inhibitor of one or both of ERK1 and ERK2 is Compound 1, or a pharmaceutically acceptable salt thereof, as described herein.
  • a TOR inhibitor is a compound of formula I, as described herein, infra.
  • a TOR inhibitor is any TOR inhibitor known to one of ordinary skill in the art. Such TOR inhibitors are described herein, infra. Definitions
  • An“alkyl” group is a saturated, partially saturated, or unsaturated straight chain or branched non-cyclic hydrocarbon having from 1 to 10 carbon atoms, typically from 1 to 8 carbons or, in some embodiments, from 1 to 6, 1 to 4, or 2 to 6 or carbon atoms.
  • Representative alkyl groups include -methyl, -ethyl, -n-propyl, -n-butyl, -n-pentyl and -n-hexyl; while saturated branched alkyls include -isopropyl, -sec-butyl, -isobutyl, -tert-butyl, -isopentyl, 2-methylpentyl, 3-methylpentyl, 4-methylpentyl, 2,3-dimethylbutyl and the like.
  • An alkyl group can be substituted or unsubstituted.
  • alkyl groups described herein when they are said to be “substituted,” they may be substituted with any substituent or substituents as those found in the exemplary compounds and embodiments disclosed herein, as well as halogen (chloro, iodo, bromo, or fluoro); hydroxyl; alkoxy; alkoxyalkyl; amino; alkylamino; carboxy; nitro; cyano; thiol; thioether; imine; imide; amidine; guanidine; enamine; aminocarbonyl; acylamino; phosphonate; phosphine; thiocarbonyl; sulfinyl; sulfone; sulfonamide; ketone; aldehyde; ester; urea; urethane; oxime; hydroxyl amine; alkoxyamine; aralkoxyamine; N-oxide; hydrazine; hydrazide; hydrazone
  • An“alkenyl” group is a straight chain or branched non-cyclic hydrocarbon having from 2 to 10 carbon atoms, typically from 2 to 8 carbon atoms, and including at least one carbon- carbon double bond.
  • Representative straight chain and branched (C 2 -C 8 )alkenyls include -vinyl, -allyl, -1-butenyl, -2-butenyl, -isobutylenyl, -1-pentenyl, -2-pentenyl, -3-methyl-1-butenyl, -2- methyl-2-butenyl, -2,3-dimethyl-2-butenyl, -1-hexenyl, -2-hexenyl, -3-hexenyl, -1-heptenyl, -2- heptenyl, -3-heptenyl, -1-octenyl, -2-octenyl, -3-octenyl and the
  • A“cycloalkyl” group is a saturated, or partially saturated cyclic alkyl group of from 3 to 10 carbon atoms having a single cyclic ring or multiple condensed or bridged rings which can be optionally substituted with from 1 to 3 alkyl groups.
  • the cycloalkyl group has 3 to 8 ring members, whereas in other embodiments the number of ring carbon atoms ranges from 3 to 5, 3 to 6, or 3 to 7.
  • Such cycloalkyl groups include, by way of example, single ring structures such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, 1-methylcyclopropyl, 2-methylcyclopentyl, 2-methylcyclooctyl, and the like, or multiple or bridged ring structures such as adamantyl and the like.
  • Examples of unsaturated cycloalkyl groups include cyclohexenyl, cyclopentenyl, cyclohexadienyl, butadienyl, pentadienyl, hexadienyl, among others.
  • a cycloalkyl group can be substituted or unsubstituted.
  • substituted cycloalkyl groups include, by way of example, cyclohexanone and the like.
  • An“aryl” group is an aromatic carbocyclic group of from 6 to 14 carbon atoms having a single ring (e.g., phenyl) or multiple condensed rings (e.g., naphthyl or anthryl).
  • aryl groups contain 6-14 carbons, and in others from 6 to 12 or even 6 to 10 carbon atoms in the ring portions of the groups.
  • Particular aryls include phenyl, biphenyl, naphthyl and the like.
  • An aryl group can be substituted or unsubstituted.
  • aryl groups also includes groups containing fused rings, such as fused aromatic-aliphatic ring systems (e.g., indanyl, tetrahydronaphthyl, and the like).
  • A“heteroaryl” group is an aryl ring system having one to four heteroatoms as ring atoms in a heteroaromatic ring system, wherein the remainder of the atoms are carbon atoms.
  • heteroaryl groups contain 5 to 6 ring atoms, and in others from 6 to 9 or even 6 to 10 atoms in the ring portions of the groups. Suitable heteroatoms include oxygen, sulfur and nitrogen.
  • the heteroaryl ring system is monocyclic or bicyclic.
  • Non-limiting examples include but are not limited to, groups such as pyrrolyl, pyrazolyl, imidazolyl, triazolyl, tetrazolyl, oxazolyl, isoxazolyl, thiazolyl, pyrrolyl, pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl, thiophenyl, benzothiophenyl, furanyl, benzofuranyl (for example, isobenzofuran-1,3-diimine), indolyl, azaindolyl (for example, pyrrolopyridyl or 1H- pyrrolo[2,3-b]pyridyl), indazolyl, benzimidazolyl (for example, 1H-benzo[d]imidazolyl), imidazopyridyl (for example, azabenzimidazolyl, 3H-imidazo[4,5-b]pyr
  • A“heterocyclyl” is an aromatic (also referred to as heteroaryl) or non-aromatic cycloalkyl in which one to four of the ring carbon atoms are independently replaced with a heteroatom from the group consisting of O, S and N.
  • heterocyclyl groups include 3 to 10 ring members, whereas other such groups have 3 to 5, 3 to 6, or 3 to 8 ring members.
  • Heterocyclyls can also be bonded to other groups at any ring atom (i.e., at any carbon atom or heteroatom of the heterocyclic ring).
  • a heterocyclylalkyl group can be substituted or unsubstituted.
  • Heterocyclyl groups encompass unsaturated, partially saturated and saturated ring systems, such as, for example, imidazolyl, imidazolinyl and imidazolidinyl groups.
  • heterocyclyl includes fused ring species, including those comprising fused aromatic and non- aromatic groups, such as, for example, benzotriazolyl, 2,3-dihydrobenzo[l,4]dioxinyl, and benzo[l,3]dioxolyl.
  • the phrase also includes bridged polycyclic ring systems containing a heteroatom such as, but not limited to, quinuclidinyl.
  • heterocyclyl group examples include, but are not limited to, aziridinyl, azetidinyl, pyrrolidyl, imidazolidinyl, pyrazolidinyl, thiazolidinyl, tetrahydrothiophenyl, tetrahydrofuranyl, dioxolyl, furanyl, thiophenyl, pyrrolyl, pyrrolinyl, imidazolyl, imidazolinyl, pyrazolyl, pyrazolinyl, triazolyl, tetrazolyl, oxazolyl, isoxazolyl, thiazolyl, thiazolinyl, isothiazolyl, thiadiazolyl, oxadiazolyl, piperidyl, piperazinyl, morpholinyl, thiomorpholinyl, tetrahydropyranyl (for example, tetrahydro-2H
  • substituted heterocyclyl groups may be mono- substituted or substituted more than once, such as, but not limited to, pyridyl or morpholinyl groups, which are 2-, 3-, 4-, 5-, or 6-substituted, or disubstituted with various substituents such as those listed below.
  • A“cycloalkylalkyl” group is a radical of the formula: -alkyl-cycloalkyl, wherein alkyl and cycloalkyl are defined above. Substituted cycloalkylalkyl groups may be substituted at the alkyl, the cycloalkyl, or both the alkyl and the cycloalkyl portions of the group. Representative cycloalkylalkyl groups include but are not limited to cyclopentylmethyl, cyclopentylethyl, cyclohexylmethyl, cyclohexylethyl, and cyclohexylpropyl. Representative substituted cycloalkylalkyl groups may be mono- substituted or substituted more than once.
  • An“aralkyl” group is a radical of the formula: -alkyl-aryl, wherein alkyl and aryl are defined above. Substituted aralkyl groups may be substituted at the alkyl, the aryl, or both the alkyl and the aryl portions of the group. Representative aralkyl groups include but are not limited to benzyl and phenethyl groups and fused (cycloalkylaryl)alkyl groups such as 4-ethyl-indanyl.
  • A“heterocyclylalkyl” group is a radical of the formula: -alkyl-heterocyclyl, wherein alkyl and heterocyclyl are defined above. Substituted heterocyclylalkyl groups may be substituted at the alkyl, the heterocyclyl, or both the alkyl and the heterocyclyl portions of the group.
  • heterocyclylalkyl groups include but are not limited to 4-ethyl- morpholinyl, 4-propylmorpholinyl, furan-2-yl methyl, furan-3-yl methyl, pyridine-3-yl methyl, (tetrahydro-2H-pyran-4-yl)methyl, (tetrahydro-2H-pyran-4-yl)ethyl, tetrahydrofuran-2-yl methyl, tetrahydrofuran-2-yl ethyl, and indol-2-yl propyl.
  • A“halogen” is chloro, iodo, bromo, or fluoro.
  • A“hydroxyalkyl” group is an alkyl group as described above substituted with one or more hydroxy groups.
  • An“alkoxy” group is -O-(alkyl), wherein alkyl is defined above.
  • An“alkoxyalkyl” group is -(alkyl)-O-(alkyl), wherein alkyl is defined above.
  • An“amine” group is a radical of the formula: -NH 2 .
  • A“hydroxyl amine” group is a radical of the formula: -N(R # )OH or -NHOH, wherein R # is a substituted or unsubstituted alkyl, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, heterocyclyl or heterocyclylalkyl group as defined herein.
  • An“alkoxyamine” group is a radical of the formula: -N(R # )O-alkyl or -NHO-alkyl, wherein R # is as defined above.
  • An“aralkoxyamine” group is a radical of the formula: -N(R # )O-aryl or -NHO-aryl, wherein R # is as defined above.
  • An“alkylamine” group is a radical of the formula: -NH-alkyl or -N(alkyl) 2 , wherein each alkyl is independently as defined above.
  • An“N-oxide” group is a radical of the formula: -N + -O-.
  • a “hydrazine” group is a radical of the formula: -N(R # )N(R # ) 2 , -NHN(R # ) 2 , -N(R # )NH(R # ) , -N(R # )NH 2 , -NHNH(R # ) 2 , or -NHNH 2 , wherein each R # is independently as defined above.
  • An“azide” group is a radical of the formula: -N 3 .
  • A“cyanate” group is a radical of the formula: -OCN.
  • A“thiocyanate” group is a radical of the formula: -SCN.
  • A“thioether” group is a radical of the formula; -S(R # ), wherein R # is as defined above.
  • a “sulfonylamino” group is a radical of the formula: -NHSO 2 (R # ) or -N(alkyl)SO 2 (R # ), wherein each alkyl and R # are defined above.
  • A“phosphine” group is a radical of the formula: -P(R # ) 2 , wherein each R # is independently as defined above. [0092] When the groups described herein, with the exception of alkyl group are said to be “substituted,” they may be substituted with any appropriate substituent or substituents.
  • substituents are those found in the exemplary compounds and embodiments disclosed herein, as well as halogen (chloro, iodo, bromo, or fluoro); alkyl; hydroxyl; alkoxy; alkoxyalkyl; amino; alkylamino; carboxy; nitro; cyano; thiol; thioether; imine; imide; amidine; guanidine; enamine; aminocarbonyl; acylamino; phosphonate; phosphine; thiocarbonyl; sulfinyl; sulfone; sulfonamide; ketone; aldehyde; ester; urea; urethane; oxime; hydroxyl amine; alkoxyamine; aralkoxyamine; N-oxide; hydrazine; hydrazide; hydrazone; azide; isocyanate; isothiocyanate; cyanate; thiocyanate
  • the term“pharmaceutically acceptable salt(s)” refers to a salt prepared from a pharmaceutically acceptable non-toxic acid or base including an inorganic acid and base and an organic acid and base.
  • Suitable pharmaceutically acceptable base addition salts include, but are not limited to metallic salts made from aluminum, calcium, lithium, magnesium, potassium, sodium and zinc or organic salts made from lysine, N,N’-dibenzylethylenediamine, chloroprocaine, choline, diethanolamine, ethylenediamine, meglumine (N-methylglucamine) and procaine.
  • Suitable non-toxic acids include, but are not limited to, inorganic and organic acids such as acetic, alginic, anthranilic, benzenesulfonic or besylate, benzoic, camphorsulfonic, citric, ethenesulfonic, formic, fumaric, furoic, galacturonic, gluconic, glucuronic, glutamic, glycolic, hydrobromic, hydrochloric, isethionic, lactic, maleic, malic, mandelic, methanesulfonic, mucic, nitric, pamoic, pantothenic, phenylacetic, phosphoric, propionic, salicylic, stearic, succinic, sulfanilic, sulfuric, tartaric acid, and p-toluenesulfonic acid.
  • inorganic and organic acids such as acetic, alginic, anthranilic, benzenesulfonic or besylate,
  • Non-toxic acids include hydrochloric, hydrobromic, phosphoric, sulfuric, and methanesulfonic acids.
  • Examples of specific salts thus include hydrochloride and mesylate salts.
  • Others are well-known in the art, see for example, Remington’s Pharmaceutical Sciences, 18 th eds., Mack Publishing, Easton PA (1990) or Remington: The Science and Practice of Pharmacy, 19 th eds., Mack Publishing, Easton PA (1995).
  • the term“clathrate” means a TOR inhibitor, 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 or a crystal lattice wherein a TOR inhibitor is a guest molecule.
  • the term“solvate” means a TOR inhibitor, or a salt thereof, that further includes a stoichiometric or non-stoichiometric amount of a solvent bound by non-covalent intermolecular forces.
  • the solvate is a hydrate.
  • the term“hydrate” means a TOR inhibitor, or a salt thereof, that further includes a stoichiometric or non-stoichiometric amount of water bound by non-covalent intermolecular forces.
  • prodrug means a TOR inhibitor derivative that can hydrolyze, oxidize, or otherwise react under biological conditions (in vitro or in vivo) to provide an active compound, particularly a TOR inhibitor.
  • prodrugs include, but are not limited to, derivatives and metabolites of a TOR inhibitor that include biohydrolyzable moieties such as biohydrolyzable amides, biohydrolyzable esters, biohydrolyzable carbamates, biohydrolyzable carbonates, biohydrolyzable ureides, and biohydrolyzable phosphate analogues.
  • prodrugs of compounds with carboxyl functional groups are the lower alkyl esters of the carboxylic acid.
  • the carboxylate esters are conveniently formed by esterifying any of the carboxylic acid moieties present on the molecule.
  • Prodrugs can typically be prepared using well-known methods, such as those described by Burger’s Medicinal Chemistry and Drug Discovery 6 th ed. (Donald J. Abraham ed., 2001, Wiley) and Design and Application of Prodrugs (H. Bundgaard ed., 1985, Harwood Academic Publishers Gmfh).
  • stereoisomer or “stereomerically pure” means one stereoisomer of a TOR inhibitor that is substantially free of other stereoisomers of that compound.
  • a stereomerically pure compound having one chiral center will be substantially free of the opposite enantiomer of the compound.
  • a stereomerically pure compound having two chiral centers will be substantially free of other diastereomers of the compound.
  • a typical stereomerically pure compound comprises greater than about 80% by weight of one stereoisomer of the compound and less than about 20% by weight of other stereoisomers of the compound, greater than about 90% by weight of one stereoisomer of the compound and less than about 10% by weight of the other stereoisomers of the compound, greater than about 95% by weight of one stereoisomer of the compound and less than about 5% by weight of the other stereoisomers of the compound, or greater than about 97% by weight of one stereoisomer of the compound and less than about 3% by weight of the other stereoisomers of the compound.
  • the TOR inhibitors can have chiral centers and can occur as racemates, individual enantiomers or diastereomers, and mixtures thereof.
  • the TOR inhibitors can include E and Z isomers, or a mixture thereof, and cis and trans isomers or a mixture thereof.
  • the TOR inhibitors are isolated as either the cis or trans isomer. In other embodiments, the TOR inhibitors are a mixture of the cis and trans isomers.
  • Tautomers refers to isomeric forms of a compound that are in equilibrium with each other. The concentrations of the isomeric forms will depend on the environment the compound is found in and may be different depending upon, for example, whether the compound is a solid or is in an organic or aqueous solution. For example, in aqueous solution, pyrazoles may exhibit the following isomeric forms, which are referred to as tautomers of each other:
  • the TOR inhibitors can contain unnatural proportions of atomic isotopes at one or more of the atoms.
  • the compounds may be radiolabeled with radioactive isotopes, such as for example tritium ( 3 H), iodine-125 ( 125 I), sulfur-35 ( 35 S), or carbon-14 ( 14 C), or may be isotopically enriched, such as with deuterium ( 2 H), carbon-13 ( 13 C), or nitrogen-15 ( 15 N).
  • an“isotopologue” is an isotopically enriched compound.
  • isotopically enriched refers to an atom having an isotopic composition other than the natural isotopic composition of that atom.“Isotopically enriched” may also refer to a compound containing at least one atom having an isotopic composition other than the natural isotopic composition of that atom.
  • isotopic composition refers to the amount of each isotope present for a given atom. Radiolabeled and isotopically enriched compounds are useful as therapeutic agents, e.g., cancer and inflammation therapeutic agents, research reagents, e.g., binding assay reagents, and diagnostic agents, e.g., in vivo imaging agents.
  • isotopologues of the TOR inhibitors are deuterium, carbon-13, or nitrogen-15 enriched TOR inhibitors.
  • a“disease or disorder associated with one or both of ERK1 and ERK2” means any disease or other deleterious condition in which one or both of ERK1 and ERK2, or a mutant thereof, is known or suspected to play a role.
  • ERK1 and ERK2 are downstream targets within the MAPK pathway.
  • a disease or disorder associated with one or both of ERK1 and ERK2 includes those in which activation of the MAPK pathway at any level (Ras-Raf-Mek-ERK) is known or suspected to play a role, including one or both of ERK1 and ERK2 as well as other nodes in the MAPK pathway upstream from ERK (such as Ras, Raf and Mek).
  • another embodiment of the present invention relates to preventing, treating, stabilizing or lessening the severity or progression of one or more diseases in which one or both of ERK1 and ERK2, or a mutant thereof, is known or suspected to play a role.
  • the present invention relates to a method of treating or lessening the severity of a proliferative disorder, wherein said method comprises administering to a patient in need thereof Compound 1 in combination with a TOR inhibitor.
  • the term“irreversible” or“irreversible inhibitor” refers to an inhibitor (i.e. a compound) that is able to be covalently bonded to a target protein kinase in a substantially non-reversible manner. That is, whereas a reversible inhibitor is able to bind to (but is generally unable to form a covalent bond to) the target protein kinase, and therefore can become dissociated from the target protein kinase, an irreversible inhibitor will remain substantially bound to the target protein kinase once covalent bond formation has occurred. Irreversible inhibitors usually display time dependency, whereby the degree of inhibition increases with the time with which the inhibitor is in contact with the enzyme.
  • Such methods include, but are not limited to, enzyme kinetic analysis of the inhibition profile of the compound with the protein kinase target, the use of mass spectrometry of the protein drug target modified in the presence of the inhibitor compound, discontinuous exposure, also known as“washout,” experiments, and the use of labeling, such as radiolabelled inhibitor, to show covalent modification of the enzyme, as well as other methods known to one of skill in the art.
  • subject means a mammal and includes human and animal subjects, such as domestic animals (e.g., horses, dogs, cats, etc.).
  • a“therapeutically effective amount” means an amount of a substance (e.g., a therapeutic agent, composition, and/or formulation) that elicits a desired biological response.
  • a therapeutically effective amount of a substance is an amount that is sufficient, when administered as part of a dosing regimen to a subject suffering from or susceptible to a disease, condition, or disorder, to treat, diagnose, prevent, and/or delay the onset of the disease, condition, or disorder.
  • the effective amount of a substance may vary depending on such factors as the desired biological endpoint, the substance to be delivered, the target cell or tissue, etc.
  • the effective amount of compound in a formulation to treat a disease, condition, or disorder is the amount that alleviates, ameliorates, relieves, inhibits, prevents, delays onset of, reduces severity of and/or reduces incidence of one or more symptoms or features of the disease, condition, or disorder.
  • a“therapeutically effective amount” is at least a minimal amount of a compound, or composition containing a compound, which is sufficient for treating one or more symptoms of a disease or disorder associated with one or both of ERK1 and ERK2.
  • the terms“treat” or“treating,” as used herein, refers to partially or completely alleviating, inhibiting, delaying onset of, preventing, ameliorating and/or relieving a disease or disorder, or one or more symptoms of the disease or disorder.
  • the terms “treatment,” “treat,” and“treating” refer to partially or completely alleviating, inhibiting, delaying onset of, preventing, ameliorating and/or relieving a disease or disorder, or one or more symptoms of the disease or disorder, as described herein.
  • treatment may be administered after one or more symptoms have developed.
  • the term “treating” includes preventing or halting the progression of a disease or disorder.
  • treatment may be administered in the absence of symptoms.
  • treatment may be administered to a susceptible individual prior to the onset of symptoms (e.g., in light of a history of symptoms and/or in light of genetic or other susceptibility factors). Treatment may also be continued after symptoms have resolved, for example to prevent or delay their recurrence.
  • the term“treating” includes preventing relapse or recurrence of a disease or disorder.
  • the expression“unit dosage form” as used herein refers to a physically discrete unit of therapeutic formulation appropriate for the subject to be treated. It will be understood, however, that the total daily usage of the compositions of the present invention will be decided by the attending physician within the scope of sound medical judgment.
  • the specific effective dose level for any particular subject or organism will depend upon a variety of factors including the disorder being treated and the severity of the disorder; activity of specific active agent employed; specific composition employed; age, body weight, general health, sex and diet of the subject; time of administration, and rate of excretion of the specific active agent employed; duration of the treatment; drugs and/or additional therapies used in combination or coincidental with specific compound(s) employed, and like factors well known in the medical arts.
  • the present invention provides methods of treating, stabilizing or lessening the severity or progression of one or more diseases or disorders associated with one or both of ERK1 and ERK2 comprising administering to a patient in need thereof an inhibitor of one or both of ERK 1 and ERK2 in combination with a TOR inhibitor.
  • the inhibitor of one or both of ERK1 and ERK2 is Compound 1 (N-(2-((2-((2-methoxy-5-methylpyridin-4-yl)amino)-5-(trifluoromethyl)pyrimidin-4-yl)amino)-5- methylphenyl)acrylamide):
  • Compound 1 is in the form of a phosphate salt.
  • Compound 1 N-(2-((2-((2-methoxy-5-methylpyridin-4-yl)amino)-5- (trifluoromethyl)pyrimidin-4-yl)amino)-5-methylphenyl)acrylamide, is designated as compound number I-90 in PCT patent application serial number PCT/US14/15256, filed February 7, 2014 and published as WO2014/124230 on August 14, 2014 (referred to herein as“the ‘230 publication,”) the entirety of which is hereby incorporated by reference. The synthesis of Compound 1 is described in detail at Example 94 of the‘230 publication.
  • Compound 1 is active in a variety of assays and therapeutic models demonstrating covalent, irreversible inhibition of one or both of ERK1 and ERK2 kinases (see, e.g., Table A of the‘230 publication).
  • the present invention provides a method of treating, stabilizing or lessening the severity or progression of one or more diseases or disorders associated with one or both of ERK1 and ERK2, comprising administering to a patient in need thereof a composition comprising Compound 1:
  • a dose or dosing regimen for a pharmaceutically acceptable salt of Compound 1 is selected from any of the doses or dosing regimens for Compound 1 as described herein.
  • Compound 1 is a potent inhibitor of the kinase activities of ERK1 and ERK2.
  • Compound 1 inhibits one or both of ERK1 and ERK2 with an IC 50 of about 10 to about 20 nM.
  • Compound 1 irreversibly inhibits ERK1 and ERK2 through formation of a covalent adduct with critical cysteine residues (amino acid 183 in ERK1 and 166 in ERK2) in the vicinity of the ATP binding pocket.
  • Compound 1 was shown to exhibit good overall kinase selectivity profile.
  • Compound 1 has demonstrated potent in vitro anti-proliferative activity against a large number of cancer cell lines of various tissue origins. Bioinformatic analyses indicate that tumors with activating mutations of BRAF are particularly sensitive to Compound 1. Notably, of 27 BRAF-mutant cancer cell lines tested, 25 (93%) demonstrated sensitivity to Compound 1 inhibition (GI50 ⁇ 1 ⁇ M). In the same cancer cell panel screening, 28 of 37 (76%) KRAS-mutant cancer cell lines were sensitive to Compound 1. Compound 1 also exhibits inhibitory activity against, for instance, A375 melanoma cells that have acquired in vitro resistance to BRAF and MEK inhibition.
  • provided methods comprise combination therapies utilizing an inhibitor of one or both of ERK1 and ERK2 and a TOR inhibitor.
  • TOR inhibitor includes mTOR kinase inhibitors and allosteric mTOR inhibitors. Examples of both mTOR kinase inhibitors and allosteric mTOR inhibitors are known to one of ordinary skill in the art and include those described herein, infra.
  • mTOR mimmalian target of rapamycin
  • FRAP FRAP
  • RAFTI RAFTI
  • RAPT1 RAFTI protein kinase
  • PI3K phosphoinositide 3-kinase
  • mTOR has been shown to be a critical protein in the PI3K/Akt/mTOR pathway that regulates cell growth and proliferation (Georgakis and Younes, Expert Rev. Anticancer Ther. 6(1):131-140 (2006)), and various mTOR inhibitors have been, or are being, evaluated in human clinical trials for the treatment of cancer.
  • the present invention provides a method of treating, stabilizing or lessening the severity or progression of one or more diseases or disorders associated with one or both of ERK1 and ERK2, or a mutant thereof, comprising administering to a patient in need thereof Compound 1, or a pharmaceutically acceptable salt thereof, in combination with a TOR inhibitor.
  • the present invention provides a method of treating, stabilizing or lessening the severity or progression of one or more diseases or disorders associated with one or both of ERK1 and ERK2, or a mutant thereof, comprising administering to a patient in need thereof Compound 1, or a pharmaceutically acceptable salt thereof, in combination with an mTOR kinase inhibitor.
  • the present invention provides a method of treating, stabilizing or lessening the severity or progression of one or more diseases or disorders associated with one or both of ERK1 and ERK2, or a mutant thereof, comprising administering to a patient in need thereof Compound 1, or a pharmaceutically acceptable salt thereof, in combination with an allosteric mTOR inhibitor.
  • Compound 1, or a pharmaceutically acceptable salt thereof is administered in combination with a TOR inhibitor of formula (I), depicted below:
  • R 1 is substituted or unsubstituted C 1-8 alkyl, substituted or unsubstituted aryl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocyclyl, or substituted or unsubstituted heterocyclylalkyl;
  • R 2 is H, substituted or unsubstituted C 1-8 alkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocyclyl, substituted or unsubstituted heterocyclylalkyl, substituted or unsubstituted aralkyl, or substituted or unsubstituted cycloalkylalkyl; and
  • R 3 is H, or a substituted or unsubstituted C 1-8 alkyl.
  • R 1 is substituted or unsubstituted aryl or substituted or unsubstituted heteroaryl.
  • R 1 is phenyl, pyridyl, pyrimidyl, benzimidazolyl, 1H-pyrrolo[2,3-b]pyridyl, indazolyl, indolyl, 1H-imidazo[4,5-b]pyridyl, 1H- imidazo[4,5-b]pyridin-2(3H)-onyl, 3H-imidazo[4,5-b]pyridyl, or pyrazolyl, each optionally substituted.
  • R 1 is phenyl substituted with one or more substituents independently selected from the group consisting of substituted or unsubstituted C 1-8 alkyl (for example, methyl), substituted or unsubstituted heterocyclyl (for example, a substituted or unsubstituted triazolyl or pyrazolyl), aminocarbonyl, halogen (for example, fluorine), cyano, hydroxyalkyl and hydroxy.
  • substituents independently selected from the group consisting of substituted or unsubstituted C 1-8 alkyl (for example, methyl), substituted or unsubstituted heterocyclyl (for example, a substituted or unsubstituted triazolyl or pyrazolyl), aminocarbonyl, halogen (for example, fluorine), cyano, hydroxyalkyl and hydroxy.
  • R 1 is pyridyl substituted with one or more substituents independently selected from the group consisting of substituted or unsubstituted C 1-8 alkyl (for example, methyl), substituted or unsubstituted heterocyclyl (for example, a substituted or unsubstituted triazolyl), halogen, aminocarbonyl , cyano, hydroxyalkyl (for example, hydroxypropyl), -OR, and -NR 2 , wherein each R is independently H, or a substituted or unsubstituted C 1-4 alkyl.
  • substituents independently selected from the group consisting of substituted or unsubstituted C 1-8 alkyl (for example, methyl), substituted or unsubstituted heterocyclyl (for example, a substituted or unsubstituted triazolyl), halogen, aminocarbonyl , cyano, hydroxyalkyl (for example, hydroxypropyl), -OR, and -NR 2
  • R 1 is 1H-pyrrolo[2,3-b]pyridyl or benzimidazolyl, optionally substituted with one or more substituents independently selected from the group consisting of substituted or unsubstituted C 1-8 alkyl, and -NR 2 , wherein R is independently H, or a substituted or unsubstituted C 1-4 alkyl.
  • R 1 is
  • R is at each occurrence independently H, or a substituted or unsubstituted C 1-4 alkyl (for example, methyl); R’ is at each occurrence independently a substituted or unsubstituted C 1-4 alkyl (for example, methyl), halogen (for example, fluoro), cyano, -OR, or - NR 2 ; m is 0-3; and n is 0-3. It will be understood by those skilled in the art that any of the subsituuents R’ may be attached to any suitable atom of any of the rings in the fused ring systems.
  • R 1 is
  • R is at each occurrence independently H, or a substituted or unsubstituted C 1-4 alkyl; R’ is at each occurrence independently a substituted or unsubstituted C 1-4 alkyl, halogen, cyano, -OR or -NR 2 ; m is 0-3; and n is 0-3.
  • R 2 is H, substituted or unsubstituted C 1-8 alkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocyclyl, substituted or unsubstituted C 1-4 alkyl-heterocyclyl, substituted or unsubstituted C 1- 4 alkyl-aryl, or substituted or unsubstituted C 1-4 alkyl-cycloalkyl.
  • R 2 is H, methyl, ethyl, n-propyl, isopropyl, n-butyl, sec-butyl, isobutyl, tert-butyl, n-pentyl, isopentyl, cyclopentyl, cyclohexyl, tetrahydrofuranyl, tetrahydropyranyl, (C1-4 alkyl)-phenyl, (C1-4 alkyl)- cyclopropyl, (C1-4 alkyl)-cyclobutyl, (C1-4 alkyl)-cyclopentyl, (C1-4 alkyl)-cyclohexyl, (C1-4 alkyl)-pyrrolidyl, (C1-4 alkyl)-piperidyl, (C1-4 alkyl)-piperazinyl, (C1-4 alkyl)-morpholinyl, (C1-4 alkyl)-tetrahydrofuranyl, or (C1-4 alkyl
  • R2 is H, C1-4 alkyl, (C1-4 alkyl)(OR),
  • R is at each occurrence independently H, or a substituted or unsubstituted C 1-4 alkyl (for example, methyl); R’ is at each occurrence independently H, -OR, cyano, or a substituted or unsubstituted C 1-4 alkyl (for example, methyl); and p is 0-3.
  • R 2 is H, C 1-4 alkyl, (C 1-4 alkyl)(OR),
  • R is at each occurrence independently H, or a substituted or unsubstituted C 1-2 alkyl; R’ is at each occurrence independently H, -OR, cyano, or a substituted or unsubstituted C 1-2 alkyl; and p is 0-1.
  • R3 is H.
  • R1 is substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl.
  • R1 is phenyl, pyridyl, pyrimidyl, benzimidazolyl, 1H-pyrrolo[2,3-b]pyridyl, indazolyl, indolyl, 1H-imidazo[4,5-b]pyridine, pyridyl, 1H-imidazo[4,5-b]pyridin-2(3H)-onyl, 3H-imidazo[4,5-b]pyridyl, or pyrazolyl, each optionally substituted.
  • R1 is phenyl substituted with one or more substituents independently selected from the group consisting of substituted or unsubstituted C1-8 alkyl, substituted or unsubstituted heterocyclyl, aminocarbonyl, halogen, cyano, hydroxyalkyl and hydroxy.
  • R1 is pyridyl substituted with one or more substituents independently selected from the group consisting of C1-8 alkyl, substituted or unsubstituted heterocyclyl, halogen, aminocarbonyl, cyano, hydroxyalkyl, -OR, and -NR2, wherein each R is independently H, or a substituted or unsubstituted C1-4 alkyl.
  • R1 is 1H- pyrrolo[2,3-b]pyridyl or benzimidazolyl, optionally substituted with one or more substituents independently selected from the group consisting of substituted or unsubstituted C1-8 alkyl, and -NR2, wherein R is independently H, or a substituted or unsubstituted C1-4 alkyl.
  • the compounds of formula (I) have an R1 group set forth herein and an R2 group set forth herein.
  • the compound at a concentration of 10 ⁇ M inhibits mTOR, DNA-PK, PI3K, or a combination thereof by at least about 50%.
  • Compounds of formula (I) may be shown to be inhibitors of the kinases above in any suitable assay system.
  • Representative TOR inhibitors of formula (I) include compounds from Table A.
  • a TOR inhibitor of Formula (I) is 7-(6-(2-hydroxypropan-2- yl)pyridin-3-yl)-1-((1r,4r)-4-methoxycyclohexyl)-3,4-dihydropyrazino-[2,3-b]pyrazin-2(1H)- one, alternatively named 7-(6-(2-hydroxypropan-2-yl)pyridin-3-yl)-1-((trans)-4- methoxycyclohexyl)-3,4-dihydropyrazino[2,3-b]pyrazin-2(1H)-one, or 7-(6-(2-hydroxypropan- 2-yl)pyridin-3-yl)-1-((1R*,4R*)-4-methoxycyclohexyl)-3,4-dihydropyrazino[2,3-b]pyrazin- 2(1H)-one, depicted below and referred to
  • a TOR inhibitor of Formula (I) is a pharmaceutically acceptable salt of Compound 2.
  • a TOR inhibitor of Formula (I) is 1 -((trans)-4- hydroxycyclohexyl)-7-(6-(2-hydroxypropan-2-yl)pyridin-3-yl)-3,4-dihydropyrazino[2,3- b]pyrazin-2(1H)-one, alternatively named 1-((1r,4r)-4-hydroxycyclohexyl)-7-(6-(2- hydroxypropan-2-yl)pyridin-3-yl)-3,4-dihydropyrazino[2,3-b]pyrazin-2(1H)-one, depicted below and referred to herein as Compound 3:
  • a TOR inhibitor of Formula (I) is a pharmaceutically acceptable salt of Compound 3.
  • a TOR inhibitor of Formula (I) is 1-ethyl-7-(2-methyl-6-(1H- 1,2,4-triazol-3-yl)pyridin-3-yl)-3,4-dihydropyrazino[2,3-b]pyrazin-2(1H)-one, or a tautomer thereof, for example, 1-ethyl-7-(2-methyl-6-(4H-1,2,4-triazol-3-yl)pyridin-3-yl)-3,4- dihydropyrazino[2,3-b]pyrazin-2(1H)-one, or 1-ethyl-7-(2-methyl-6-(1H-1,2,4-triazol-5- yl)pyridin-3-yl)-3,4-dihydropyrazino[2,3-b]pyrazin-2(1H)-one.
  • One such tautomer is depicted below and referred to herein as Compound 4:
  • a TOR inhibitor of Formula (I) is a pharmaceutically acceptable salt of Compound 4.
  • TOR inhibitors can be prepared via standard, well-known synthetic methodology, see e.g., March, J. Advanced Organic Chemistry; Reactions Mechanisms, and Structure, 4th ed., 1992. Starting materials useful for preparing compounds of formula (I) and intermediates therefore, are commercially available or can be prepared from commercially available materials using known synthetic methods and reagents.
  • a TOR inhibitor is selected from rapamycin or an analog thereof.
  • a rapamycin analog in the form of an ester, ether, hydrazone, hydroxylamine, or oxime derivatives of rapamycin.
  • a TOR inhibitor is AP23573 (Deforolimus), AP-23675, AP-23841, ABT-578 (Zotarolimus), CCI779 (Temsirolimus), RAD-001 (Everolimus), 7-epi-rapamycin, 7-thiomethyl-rapamycin, 7-epi- trimethoxyphenyl-rapamycin, 7-epi-thiomethyl-rapamycin, and 7-demethoxy-rapamycin, 32- demethoxy-rapamycin, 2-desmethyl-rapamycin, 42-O-(2-hydroxy)ethyl rapamycin, Ridaforolimus, AZD8055, OSI-027, INK-128 (MLN0128), wortmannin, and LY29004.
  • a TOR inhibitor is selected from Everolimus, Temsirolimus, Ridaforolimus, AZD8055, OSI-027, INK-128 (MLN0128), wortmannin, and LY29004.
  • a TOR inhibitor is a compound having the following structure:
  • o r a pharmaceutically acceptable salt, clathrate, solvate, stereoisomer, tautomer, or prodrug thereof.
  • a TOR inhibitor is a compound having the following structure:
  • a TOR inhibitor is a compound having the following structure:
  • a TOR inhibitor is a compound having the following structure:
  • a TOR inhibitor is a compound having the following structure:
  • o r a pharmaceutically acceptable salt, clathrate, solvate, stereoisomer, tautomer, or prodrug thereof.
  • a TOR inhibitor is a compound having the following structure:
  • o r a pharmaceutically a utomer, or prodrug thereof.
  • a TOR inhibitor is a compound having the following structure:
  • o r a pharmaceutically acceptable salt, clathrate, solvate, stereoisomer, tautomer, or prodrug thereof.
  • a TOR inhibitor is a compound having the following structure:
  • a TOR inhibitor is a compound having the following structure:
  • a TOR inhibitor is a compound disclosed in the patent publications listed below in Table B.
  • WO 2008/023161 see, e.g., page 5, line 5 to page 11, line 15
  • WO 2009/007751 see, e.g., page 9, line 8 to page 26, line 8
  • WO 2009/007749 see, e.g., page 9, line 21 to page 29, line 23
  • WO 2009/007750 see, e.g., page 9, line 21 to page 32, line 22
  • WO 2009/007748 see, e.g., page 9, line 6 to page 42, line 28
  • WO 2008/032028 see, e.g., page 11, line 13 to page 21, line 13
  • WO 2008/032086 see, e.g., page 10 line 21 to page 15, line 22
  • WO 2008/032072 see, e.g., page 11, line 11 to page 16, line 13
  • WO 2008/032033 see, e.g., page 11, line 3 to page 16, line 5
  • WO 2008/032089 see, e.g.
  • Compound 1, and pharmaceutically acceptable salts thereof described herein is an inhibitor of one or both of ERK1 and ERK2.
  • ERK is one of the key components in the RAS-RAF-MEK-ERK MAPK pathway and that ERK1 and ERK2 are downstream nodes within the MAPK pathway.
  • an ERK inhibitor can treat disease or disorders in which activation of the MAPK pathway at any level (Ras-Raf-Mek-ERK) is known or suspected to play a role, including one or both of ERK1 and ERK2 as well as other nodes in the MAPK pathway upstream from ERK (such as Ras, Raf and Mek).
  • ERK is a downstream target
  • ERK inhibitors are believed to be able to overcome, in some instances, drug resistance induced by inhibitors of targets upstream of ERK within the MAPK pathway.
  • RAF or MEK utilized in the treatment of K-RAS and B-RAF mutant tumors have resulted in such drug resistance.
  • drug resistance has been associated with other tumors driven by hyperactivation of the MAPK pathway (such as NF1 mutant tumors).
  • Kinase selectivity was achieved through silencing the selective Cys in a combination of the interactions between the covalent inhibitors of the invention and unique amino acids in the ATP binding pocket. Targeting the selective Cys provides for prolonged pharmacodynamics in silencing ERK activity, as well as potential lower doses in cancer treatment, compared to reversible inhibitors.
  • the activity of Compound 1, and pharmaceutically acceptable salts thereof, as an inhibitor of one or both of an ERK1 and ERK2 kinase, or a mutant thereof, may be assayed in vitro, in vivo or in a cell line.
  • In vitro assays include assays that determine inhibition of downstream phosphorylation, changes in gene expression, subsequent functional markers and consequences, and/or kinase activity of one or both of activated ERK1 and ERK2 kinase, or a mutant thereof. Alternate in vitro assays quantitate the ability of the test compound to bind to one or both of ERK1 and ERK2.
  • Test compound binding may be measured by radiolabeling the test compound prior to binding, isolating one or both of the compound / ERK1 complex and the compound / ERK2 complex, and determining the amount of radiolabel bound.
  • test compound binding may be determined by running a competition experiment where test compounds are incubated with one or both of ERK1 and ERK2 kinase bound to known radioligands.
  • Test compound binding may be determined by competition with an ERK covalent probe that is amenable to further functionalization with a detection probe, such as, for example, a fluorophore, biotin conjugate, radiolabel, or any other probe that facilitates its quantification.
  • a detection probe such as, for example, a fluorophore, biotin conjugate, radiolabel, or any other probe that facilitates its quantification.
  • a detection probe such as, for example, a fluorophore, biotin conjugate, radiolabel, or any other probe that facilitates its quantification
  • the term "measurably inhibit”, as used herein means a measurable change in one or both of ERK1 and ERK2 protein kinase activity between a sample comprising a provided composition, and one or both of an ERK1 and ERK2 protein kinase and an equivalent sample comprising one or both of ERK1 and ERK2 protein kinase in the absence of a provided composition.
  • Such measurements of protein kinase activity are known to one of ordinary skill in the art and include those methods set forth herein below and/or in the Examples of the‘230 publication.
  • Compound 1, and pharmaceutically acceptable salts thereof are inhibitors of one or both of ERK1 and ERK2 protein kinases, and ERK1 and ERK2 are downstream targets within the MAPK pathway.
  • ERK1 and ERK2 are inhibitors of one or both of ERK1 and ERK2 protein kinases, and ERK1 and ERK2 are downstream targets within the MAPK pathway.
  • such compounds and compositions are particularly useful for treating or lessening the severity of a disease, condition, or disorder in which activation of the MAPK pathway at any level (Ras-Raf-Mek-ERK) is known or suspected to play a role.
  • Such disease, condition, or disorder may be referred to herein as associated with the MAPK pathway or alternatively as associated with one or both of ERK1 and ERK2.
  • Such diseases, conditions, or disorders may also be refererd to herein as an "ERK1- or ERK2-mediated disease, condition, or disorder.”
  • the present invention provides a method for treating or lessening the severity of a disease, condition, or disorder where activation of the MAPK pathway (at any level in Ras-Raf-Mek-ERK), including one or both of ERK1 and ERK2 protein kinases, is implicated in said disease, condition, or disorder wherein said method comprises administering to a patient in need thereof Compound 1, or a pharmaceutically acceptable salt thereof, in combination with a TOR inhibitor.
  • the present invention relates to a method of inhibiting one or both of ERK1 and ERK2 protein kinase activity in a patient comprising the step of administering to said patient a composition comprising Compound 1, or a pharmaceutically acceptable salt thereof, in combination with a TOR inhibitor, or a composition comprising any of the foregoing.
  • the present invention provides a method for treating a disease, condition, or disorder mediated by one or both of ERK1 and ERK2 kinase, or a mutant thereof, in a patient in need thereof, comprising the step of administering to said patient Compound 1, or pharmaceutically acceptable salts thereof, in combination with a TOR inhibitor, or a pharmaceutically acceptable composition comprising any of the foregoing.
  • a disease, condition, or disorder mediated by one or both of ERK1 and ERK2 kinase, or a mutant thereof in a patient in need thereof, comprising the step of administering to said patient Compound 1, or pharmaceutically acceptable salts thereof, in combination with a TOR inhibitor, or a pharmaceutically acceptable composition comprising any of the foregoing.
  • the present invention provides a method for overcoming drug resistance to Raf or MEK inhibitors, comprising the step of administering to a patient an inhibitor compound of one or both of ERK1 and ERK2, such as Compound 1, or a pharmaceutically acceptable salt thereof, either alone or in combination with a Tor inhibitor.
  • the mechanism of drug resistance is through mutation of a target protein or reactivation of the MAPK pathway.
  • the term“resistance” may refer to changes in a wild-type nucleic acid sequence coding a target protein, and/or to the amino acid sequence of the target protein and/or to the amino acid sequence of another protein, which changes, decreases or abolishes the inhibitory effect of the inhibitor on the target protein.
  • the term“resistance” may also refer to overexpression or silencing of a protein differing from a target protein that can reactivate the MAPK pathway or other survival pathways.
  • treatment is administered after one or more symptoms have developed. In other embodiments, treatment is administered in the absence of symptoms. For example, treatment is administered to a susceptible individual prior to the onset of symptoms (e.g., in light of a history of symptoms and/or in light of genetic or other susceptibility factors). Treatment is also continued after symptoms have resolved, for example to prevent, delay or lessen the severity of their recurrence.
  • the present invention provides a method of treating, stabilizing or lessening the severity or progression of one or more diseases or disorders associated with one or both of ERK1 and ERK2 comprising administering to a patient in need thereof a composition comprising Compound 1, or a pharmaceutically acceptable salt thereof, in combination with a TOR inhibitor of Formula (I).
  • the present invention provides a method of treating, stabilizing or lessening the severity or progression of one or more diseases or disorders associated with one or both of ERK1 and ERK2 comprising administering to a patient in need thereof a composition comprising Compound 1, or a pharmaceutically acceptable salt thereof, in combination with a compound selected from Compound 2, Compound 3, or Compound 4, or a pharmaceutically acceptable salt thereof.
  • the present invention provides a method of treating, stabilizing or lessening the severity or progression of one or more diseases or disorders associated with one or both of ERK1 and ERK2 comprising administering to a patient in need thereof a composition comprising Compound 1, or a pharmaceutically acceptable salt thereof, in combination with Compound 2, or a pharmaceutically acceptable salt thereof.
  • the present invention provides a method of treating, stabilizing or lessening the severity or progression of one or more diseases or disorders associated with one or both of ERK1 and ERK2 comprising administering to a patient in need thereof a composition comprising Compound 1, or a pharmaceutically acceptable salt thereof, in combination with Compound 3, or a pharmaceutically acceptable salt thereof.
  • the present invention provides a method of treating, stabilizing or lessening the severity or progression of one or more diseases or disorders associated with one or both of ERK1 and ERK2 comprising administering to a patient in need thereof a composition comprising Compound 1, or a pharmaceutically acceptable salt thereof, in combination with Compound 4, or a pharmaceutically acceptable salt thereof.
  • the present invention provides a system for treating, stabilizing or lessening the severity of one or more diseases or disorders associated with one or more of ERK1 and ERK2, the system comprising Compound 1, or a pharmaceutically acceptable salt thereof, in combination with a TOR inhibitor.
  • the present invention contemplates a system comprising any of the above-described TOR inhibitors.
  • a TOR inhibitor is selected from rapamycin, AP23573 (Deforolimus), AP-23675, AP-23841, ABT-578 (Zotarolimus), CCI779 (Temsirolimus), RAD- 001 (Everolimus), 7-epi-rapamycin, 7-thiomethyl-rapamycin, 7-epi-trimethoxyphenyl- rapamycin, 7-epi-thiomethyl-rapamycin, and 7-demethoxy-rapamycin, 32-demethoxy- rapamycin, 2-desmethyl-rapamycin, 42-O-(2-hydroxy)ethyl rapamycin, Ridaforolimus, AZD8055, OSI-027, INK-128, MLN0128, wortmannin, and LY29004.
  • the present invention provides a system for treating, stabilizing or lessening the severity of one or more diseases or disorders associated with one or more of ERK1 and ERK2, the system comprising Compound 1, or a pharmaceutically acceptable salt thereof, in combination with a TOR inhibitor, wherein the TOR inhibitor is a compound of Formula (I).
  • a TOR inhibitor is selected from any of Compound 2, Compound 3, or Compound 4, or a pharmaceutically acceptable salt thereof.
  • General diseases, conditions, or disorders treated by Compound 1, and pharmaceutically acceptable salts thereof, in combination with a TOR inhibitor include cancer, an autoimmune disorder, a neurodegenerative or neurological disorder, liver disease, a cardiac disorder, schizophrenia, or a bone-related disorder.
  • the present invention provides a method for treating an ERK1- or ERK2-mediated disease, condition, or disorder comprising administering to a patient in need thereof Compound 1, or a pharmaceutically acceptable salt thereof, in combination with a TOR inhibitor.
  • the present invention relates to a method of treating or lessening the severity of a disease, condition, or disorder selected from cancer, stroke, diabetes, hepatomegaly, cardiovascular disease including cardiomegaly, Alzheimer's disease, cystic fibrosis, viral disease, autoimmune diseases, atherosclerosis, restenosis, psoriasis, allergic disorders including asthma, inflammation, neurological disorders and hormone-related diseases, wherein the method comprises administering to a patient in need thereof a composition comprising Compound 1, or a pharmaceutically acceptable salt thereof, in combination with a TOR inhibitor.
  • the present invention relates to a method of treating a cancer wherein the method comprises administering to a patient in need thereof a composition comprising Compound 1, or a pharmaceutically acceptable salt thereof, in combination with a TOR inhibitor.
  • the cancer is recurring.
  • the cancer is refractory.
  • the cancer is metastatic.
  • the cancer is locally advanced.
  • the cancer is a RAF inhibitor-resistant cancer.
  • the RAF inhibitor-resistant cancer is a BRAF inhibitor-resistant cancer.
  • the cancer is a MEK inhibitor-resistant cancer.
  • the cancer is a MAPK pathway-mediated cancer.
  • the cancer is a BRAF-mutated cancer.
  • the BRAF-mutated cancer is a BRAF V600 -mutated cancer, such as BRAF V600E BRAF V600K , BRAF V600R , and BRAF V600D .
  • the cancer is a RAS-mutated cancer.
  • the RAS-mutated involves codons 12, 13, or 61.
  • the RAS-mutated cancer is a KRAS-mutated cancer, including, but not limited to, KRAS G12C/D/V , KRAS G13C/D ,or KRAS Q61L/H/R .
  • the RAS-mutated cancer is an NRAS-mutated cancer, including, but not limited to, NRAS Q61R , NRAS Q61K , NRAS Q61L , or NRAS Q61H .
  • the RAS-mutated cancer is an HRAS-mutated cancer, including, but not limited to, HRAS G12V , HRAS Q61R , and HRAS G12S .
  • the present invention relates to a method of treating a cancer wherein the method comprises administering to a patient in need thereof a composition comprising Compound 1, or a pharmaceutically acceptable salt thereof, in combination with a TOR inhibitor, wherein the cancer is selected from multiple myeloma, breast, ovary, cervix, prostate, testis, genitourinary tract, esophagus, larynx, glioblastoma, neuroblastoma, stomach (gastric), skin, keratoacanthoma, lung, epidermoid carcinoma, large cell carcinoma, small cell carcinoma, lung, bone, colon, thyroid, adenoma, pancreas, adenocarcinoma, thyroid, follicular carcinoma, undifferentiated carcinoma, papillary carcinoma, seminoma, melanoma (including uveal melanoma) sarcoma, bladder carcinoma, liver carcinoma (e.g., hepatocellular carcinoma (HCC)
  • the present invention relates to a method of treating a cancer wherein the method comprises administering to a patient in need thereof a composition comprising Compound 1, or a pharmaceutically acceptable salt thereof, in combination with a TOR inhibitor, wherein the cancer is selected from carcinoma, lymphoma, blastoma, sarcoma, and leukemia.
  • a sarcoma is a soft tissue sarcoma.
  • a lymphoma is non-hodgkins lymphoma.
  • a lymphoma is large cell immunoblastic lymphoma.
  • the cancer is selected from adenocarcinoma; adenoma; adrenocortical cancer; bladder cancer; bone cancer; brain cancer; breast cancer; cancer of the buccal cavity; cervical cancer; colon cancer; colorectal cancer; endometrial or uterine carcinoma; epidermoid carcinoma; esophogeal cancer; eye cancer; follicular carcinoma; gallbladder cancer; prostate, AML, multiple myeloma (MM), gastrointestinal cancer, such as, for example, gastrointestinal stromal tumor; cancer of the genitourinary tract; glioblastoma; hairy cell carcinoma; various types of head and neck cancer; hepatic carcinoma; hepatocellular cancer; Hodgkin's disease; keratoacanthoma; kidney cancer; large cell carcinoma; cancer of the large intestine; laryngeal cancer; liver cancer; lung cancer, such as, for example, adenocarcinoma of the lung, anaplastic carcinoma of the lung,
  • the present invention relates to a method of treating a cancer wherein the method comprises administering to a patient in need thereof a composition comprising Compound 1, or a pharmaceutically acceptable salt thereof, in combination with a TOR inhibitor, wherein the cancer is selected from melanoma, pancreatic cancer, thyroid cancer, colorectal cancer, lung cancer (e.g., non-small cell lung cancer), breast cancer, endometrial cancer, prostate cancer, ovarian cancer, hepatocellular carcinoma (HCC), multiple myeloma (MM), and leukemia.
  • a leukemia is an acute leukemia.
  • a leukemia is acute myeloid leukemia.
  • a leukemia is acute lymphoblastic leukemia.
  • the present invention relates to a method of treating a cancer wherein the method comprises administering to a patient in need thereof a composition comprising Compound 1, or a pharmaceutically acceptable salt thereof, in combination with a TOR inhibitor, wherein the cancer is selected from melanoma, colorectal cancer, lung cancer, or pancreatic.
  • the present invention relates to a method of treating a cancer wherein the method comprises administering to a patient in need thereof a composition comprising Compound 1, or a pharmaceutically acceptable salt thereof, in combination with a TOR inhibitor, wherein the cancer is melanoma.
  • the melanoma is uveal melanoma.
  • the melanoma is a melanoma of the skin.
  • the melanoma is locally advanced.
  • the melanoma is metastatic.
  • the melanoma is recurring.
  • the melanoma is BRAF v600 -mutated melanoma.
  • the melanoma is a RAS- mutated melanoma. In some embodiments, the melanoma is NRAS-mutated melanoma. In certain embodiments, the melanoma is wild type for KRAS, NRAS or BRAF. In certain embodiments, the melanoma is a BRAF inhibitor-resistant (e.g., vemurfenib-resistant, dabrafenib-resistant, encorafenib-resistant, etc.) melanoma. In certain embodiments, the cancer is a VemR (i.e., Vemurfenib-resistant) BRAF-mutated melanoma. In some embodiments, the melanoma is relapsed. In some embodiments, the melanoma is refractory.
  • the present invention relates to a method of treating a cancer wherein the method comprises administering to a patient in need thereof a composition comprising Compound 1, or a pharmaceutically acceptable salt thereof, in combination with a TOR inhibitor, wherein the cancer is colorectal cancer.
  • the colorectal cancer is locally advanced.
  • the colorectal cancer is metastatic.
  • the colorectal cancer is a BRAF-mutated colorectal cancer.
  • the colorectal cancer is a BRAF v600 -mutated colorectal cancer.
  • the colorectal cancer is a RAS-mutated colorectal cancer.
  • the colorectal cancer is a KRAS-mutated colorectal cancer. In certain embodiments, the colorectal cancer is a NRAS-mutated colorectal cancer. In some embodiments, the colorectal cancer is relapsed. In some embodiments, the colorectal cancer is refractory.
  • the present invention relates to a method of treating a cancer wherein the method comprises administering to a patient in need thereof a composition comprising Compound 1, or a pharmaceutically acceptable salt thereof, in combination with a TOR inhibitor, wherein the cancer is pancreatic cancer.
  • the pancreatic cancer is locally advanced.
  • the pancreatic cancer is metastatic.
  • the pancreatic cancer is a pancreatic ductal adenocarcinoma (PDAC).
  • the pancreatic cancer is a RAS-mutated pancreatic cancer.
  • the pancreatic cancer is a KRAS-mutated pancreatic cancer.
  • the pancreatic cancer is KRAS-mutated pancreatic cancer, including, but not limited to, KRAS G12C/D/V , KRAS G13C/D ,or KRAS Q61L/H/R .
  • the pancreatic cancer is relapsed.
  • the pancreatic cancer is refractory.
  • the present invention relates to a method of treating a cancer wherein the method comprises administering to a patient in need thereof a composition comprising Compound 1, or a pharmaceutically acceptable salt thereof, in combination with a TOR inhibitor, wherein the cancer is a papillary thyroid cancer.
  • the papillary thyroid cancer is locally advanced.
  • the papillary thyroid cancer is metastatic.
  • the papillary thyroid cancer is recurring.
  • the papillary thyroid cancer is BRAF-mutated papillary thyroid cancer.
  • the papillary thyroid cancer is BRAF v600 -mutated papillary thyroid cancer.
  • the papillary thyroid cancer is relapsed.
  • the papillary thyroid cancer is refractory.
  • the papillary thyroid cancer includes undifferentiated or dedifferentiated histology.
  • the present invention relates to a method of treating a cancer wherein the method comprises administering to a patient in need thereof a composition comprising Compound 1, or a pharmaceutically acceptable salt thereof, in combination with a TOR inhibitor, wherein the cancer is lung cancer.
  • the lung cancer is non-small cell lung cancer (NSCLC).
  • the lung cancer is locally advanced.
  • the lung cancer is metastatic.
  • the lung cancer is a RAS-mutated lung cancer.
  • the lung cancer is KRAS- mutated lung cancer.
  • the lung cancer is a KRAS-mutated lung cancer, including, but not limited to, KRAS G12C/D/V , KRAS G13C/D ,or KRAS Q61L/H/R .
  • the lung cancer is relapsed.
  • the lung cancer is refractory.
  • the present invention relates to a method of treating a cancer wherein the method comprises administering to a patient in need thereof a composition comprising Compound 1, or a pharmaceutically acceptable salt thereof, in combination with a TOR inhibitor, wherein the cancer is a leukemia.
  • a leukemia is a chronic leukemia.
  • a leukemia is chronic myeloid leukemia.
  • a leukemia is an acute leukemia.
  • a leukemia is acute myeloid leukemia (AML).
  • a leukemia is acute monocytic leukemia (AMoL, or AML-M5).
  • a leukemia is acute lymphoblastic leukemia (ALL). In certain embodiments, a leukemia is acute T cell leukemia. In certain embodiments, a leukemia is myelomonoblastic leukemia. In certain embodiments, a leukemia is human B cell precursor leukemia. In certain embodiments, a leukemia has a Flt3 mutation or rearrangement. In some embodiments, the leukemia is relapsed. In some embodiments, the leukemia is refractory.
  • ALL acute lymphoblastic leukemia
  • a leukemia is acute T cell leukemia.
  • a leukemia is myelomonoblastic leukemia.
  • a leukemia is human B cell precursor leukemia.
  • a leukemia has a Flt3 mutation or rearrangement. In some embodiments, the leukemia is relapsed. In some embodiments, the leukemia is refractory.
  • the present invention relates to a method of treating a cancer wherein the method comprises administering to a patient in need thereof a composition comprising Compound 1, or a pharmaceutically acceptable salt thereof, in combination with a TOR inhibitor, wherein the cancer is a CNS cancer, for instance CNS tumors.
  • a CNS tumor is a glioblastoma or glioblastoma multiforme (GBM).
  • GBM glioblastoma multiforme
  • the present invention relates to a method of treating stomach (gastric) and esophageal tumors and cancers.
  • the present invention relates to a method of treating a cancer wherein the method comprises administering to a patient in need thereof a composition comprising Compound 1, or a pharmaceutically acceptable salt thereof, in combination with a TOR inhibitor, wherein the cancer is multiple myeloma (MM).
  • the multiple myeloma is locally advanced.
  • the multiple myeloma is metastatic.
  • the multiple myeloma is a RAS-mutated multiple myeloma.
  • the multiple myeloma is KRAS-mutated multiple myeloma.
  • the multiple myeloma is a KRAS-mutated multiple myeloma, including, but not limited to, KRAS G12C/D/V , KRAS G13C/D , or KRAS Q61L/H/R .
  • the multiple myeloma is relapsed. In some embodiments, the multiple myeloma is refractory.
  • the present invention relates to a method of treating a cancer, wherein the method comprises administering to a patient in need thereof a composition comprising Compound 1, or a pharmaceutically acceptable salt thereof, in combination with a TOR inhibitor, wherein the cancer is hepatocellular carcinoma (HCC).
  • HCC hepatocellular carcinoma
  • the HCC is locally advanced.
  • the HCC is metastatic.
  • the HCC is a RAS-mutated HCC.
  • the HCC is KRAS- mutated HCC.
  • the HCC is a KRAS-mutated HCC, including, but not limited to, KRAS G12C/D/V , KRAS G13C/D , or KRAS Q61L/H/R .
  • the hepatocellular carcinoma is relapsed. In some embodiments, the hepatocellular carcinoma is refractory.
  • the present invention relates to a method of treating a cancer wherein the method comprises administering to a patient in need thereof a composition comprising Compound 1, or a pharmaceutically acceptable salt thereof, in combination with a TOR inhibitor, wherein the cancer is selected from breast, colorectal, endometrial, hematological, leukemia (e.g., AML), liver, lung, melanoma, ovarian, pancreatic, prostate, or thyroid.
  • a composition comprising Compound 1, or a pharmaceutically acceptable salt thereof, in combination with a TOR inhibitor
  • the cancer is selected from breast, colorectal, endometrial, hematological, leukemia (e.g., AML), liver, lung, melanoma, ovarian, pancreatic, prostate, or thyroid.
  • the present invention relates to a method of treating a cancer wherein the method comprises administering to a patient in need thereof a composition comprising Compound 1, or a pharmaceutically acceptable salt thereof, in combination with a TOR inhibitor, wherein the cancer is selected from breast, colorectal, endometrial, liver, lung, melanoma, ovarian, pancreatic, or thyroid.
  • the present invention relates to a method of treating a cancer wherein the method comprises administering to a patient in need thereof a composition comprising Compound 1, or a pharmaceutically acceptable salt thereof, in combination with a TOR inhibitor, wherein the cancer is selected from colorectal, lung, melanoma, or pancreatic.
  • the present invention relates to a method of treating a cancer wherein the method comprises administering to a patient in need thereof a composition comprising Compound 1, or a pharmaceutically acceptable salt thereof, in combination with a TOR inhibitor, wherein the cancer is selected from colorectal, melanoma, or pancreatic.
  • provided methods comprise administration to a patient in need thereof Compound 1, or a pharmaceutically acceptable salt thereof, in combination with a TOR inhibitor.
  • the term“in combination” with regard to administration of Compound 1 and a TOR inhibitor means that each of Compound 1 and the TOR inhibitor can be administered to the patient in any order (i.e., simultaneously or sequentially) or together in a single composition, formulation, or unit dosage form.
  • Compound 1 or a pharmaceutically acceptable salt thereof, and the TOR inhibitor can be administered on the same day or on different days and in any order as according to an appropriate dosing protocol. Dosing of Compound 1
  • the present invention provides a method of treating, stabilizing or lessening the severity or progression of one or more diseases or disorders associated with one or both of ERK1 and ERK2 comprising administering to a patient in need thereof a TOR inhibitor in combination with a particular total daily dose of Compound 1, wherein the total daily dose of Compound 1 is selected from about 5 mg, about 10 mg, about 15 mg, about 20 mg, about 25 mg, about 30 mg, about 35 mg, about 40 mg, about 45 mg, about 50 mg, about 55 mg, about 60 mg, about 65 mg, about 70 mg, about 75 mg, about 80 mg, about 85 mg, about 90 mg, about 95 mg, about 100 mg, about 105 mg, about 110 mg, about 115 mg, about 120 mg, about 125 mg, about 130 mg, about 135 mg, about 140 mg, about 145 mg, about 150 mg, about 155 mg, about 160 mg, about 165 mg, about 170 mg, about 175 mg, about 180 mg, about 185 mg, about 190 mg
  • the present invention provides a method of treating, stabilizing or lessening the severity or progression of one or more diseases or disorders associated with one or both of ERK1 and ERK2 comprising administering to a patient in need thereof a TOR inhibitor in combination with a particular total daily dose of Compound 1, wherein the total daily dose of Compound 1 is selected from about 100 mg, about 150 mg, about 200 mg, about 250 mg, about 300 mg, about 350 mg, about 400 mg, about 450 mg, about 500 mg, about 550 mg, about 600 mg, about 650 mg, about 700 mg, about 750 mg, about 800 mg, about 850 mg, about 900 mg, about 950 mg, about 1000 mg, about 1050 mg, about 1100 mg, about 1150 mg, about 1200 mg, about 1250 mg, about 1300 mg, about 1350 mg, about 1400 mg, about 1450 mg, about 1500 mg, about 1550 mg, about 1600 mg, about 1650 mg, about 1700 mg, about 1750 mg, about 1800 mg, about 18
  • the present invention provides a method of treating, stabilizing or lessening the severity or progression of one or more diseases or disorders associated with one or both of ERK1 and ERK2 comprising administering to a patient in need thereof a TOR inhibitor in combination with a particular total daily dose of Compound 1, wherein the total daily dose of Compound 1 is selected from about 10 mg to about 500 mg, or about 10 mg to about 450 mg, or about 10 mg to about 425 mg, or about 10 mg to about 400 mg, or about 10 mg to about 375 mg, or about 10 mg to about 350 mg, or about 10 mg to about 325 mg, or about10 mg to about 300 mg, or about 10 mg to about 275 mg, or about 10 to about 250 mg, or about 10 to about 225 mg, or about 10 mg to about 200 mg, or about 10 mg to about 190 mg, or about 10 mg to about 180 mg, or about 10 mg to about 170 mg, or about 10 mg to about 160 mg, or about 10 mg to about 150 mg, or about 10 mg
  • the present invention provides a method of treating, stabilizing or lessening the severity or progression of one or more diseases or disorders associated with one or both of ERK1 and ERK2 comprising administering to a patient in need thereof a TOR inhibitor in combination with a particular total daily dose of Compound 1, wherein the total daily dose of Compound 1 is selected from about 100 mg to about 3000 mg, or about 500 mg to about 3000 mg, or about 100 mg to about 2500 mg, or about 500 mg to about 2500 mg, or about 100 mg to about 2200 mg, or about 500 mg to about 2200 mg, or about 600 mg to about 2200 mg, or about 700 mg to about 2200 mg, or about 800 to about 2200 mg, or about 800 to about 2100 mg, or about 800 to about 2000 mg.
  • the daily dose is about 800 mg to about 2000 mg.
  • a total daily dose of Compound 1 is administered once daily (QD), wherein the dose is selected from about 5 mg, about 10 mg, about 20 mg, about 40 mg, about 80 mg, about 120 mg, about 180 mg, about 330 mg, about 480 mg, or about 640 mg.
  • a total daily dose of Compound 1 is administered once daily (QD), wherein the dose is selected from about 20 mg, about 40 mg, about 80 mg, or about 160 mg.
  • QD once daily
  • a dose of a TOR inhibitor to be administered to a patient is rather widely variable and can be patient to the judgment of a health-care practitioner.
  • TOR inhibitors can be administered one to four times a day in a dose of about 0.005 mg/kg of a patient’s body weight to about 10 mg/kg of a patient’s body weight in a patient, but the above dosage may be properly varied depending on the age, body weight and medical condition of the patient and the type of administration.
  • the dose is about 0.01 mg/kg of a patient’s body weight to about 5 mg/kg of a patient’s body weight, about 0.05 mg/kg of a patient’s body weight to about 1 mg/kg of a patient’s body weight, about 0.1 mg/kg of a patient’s body weight to about 0.75 mg/kg of a patient’s body weight or about 0.25 mg/kg of a patient’s body weight to about 0.5 mg/kg of a patient’s body weight.
  • one dose is given per day
  • two doses are given per day.
  • the amount of TOR inhibitor administered will depend on such factors as the solubility of the active component, the formulation used and the route of administration.
  • the present invention provides methods for treating, stabilizing or lessening the severity or progression of one or more diseases or disorders associated with one or both of ERK1 and ERK2, comprising administering to a patient in need thereof a composition comprising Compound 1, or a pharmaceutically acceptable salt thereof, and a TOR inhibitor, wherein the TOR inhibitor is administered in an amount of about 0.1 mg/day to about 1200 mg/day, about 1 mg/day to about 100 mg/day, about 10 mg/day to about 1200 mg/day, about 10 mg/day to about 100 mg/day, about 100 mg/day to about 1200 mg/day, about 400 mg/day to about 1200 mg/day, about 600 mg/day to about 1200 mg/day, about 400 mg/day to about 800 mg/day or about 600 mg/day to about 800 mg/day.
  • methods disclosed herein comprise the administration of about 0.1 mg/day, about 0.5 mg/day, about 1 mg/day, about 10 mg/day, about 15 mg/day, about 20 mg/day, about 30 mg/day, about 40 mg/day, about 45 mg/day, about 50 mg/day, about 60 mg/day, about 75 mg/day, about 100 mg/day, about 125 mg/day, about 150 mg/day, about 200 mg/day, about 250 mg/day, about 300 mg/day, about 400 mg/day, about 600 mg/day or about 800 mg/day of a TOR inhibitor to a patient in need thereof.
  • methods disclosed herein comprise the administration of about 10 mg/day, about 15 mg/day, about 20 mg/day, about 30 mg/day or about 45 mg/day.
  • methods disclosed herein comprise the administration of about 10 mg/day, about 15 mg/day, about 16 mg/day, about 20 mg/day, about 30 mg/day.
  • Unit Dosage Forms of Compound 1
  • Compound 1, or a pharmaceutically acceptable salt thereof is preferably formulated in unit dosage form for ease of administration and uniformity of dosage. It will be understood, however, that the total daily usage of Compound 1, or a pharmaceutically acceptable salt thereof, and compositions thereof, will be decided by the attending physician within the scope of sound medical judgment.
  • the specific effective dose level for any particular patient or organism will depend upon a variety of factors including the disorder being treated and the severity of the disorder; the specific composition employed; the age, body weight, general health, sex and diet of the patient; the time of administration, route of administration, and rate of excretion of Compound 1; the duration of the treatment; drugs used in combination or coincidental with Compound 1, and like factors well known in the medical arts.
  • the unit dosage forms described herein refer to an amount of Compound 1, i.e. the free base form of the active pharmaceutical ingredient, which may be provided as the free base or as a pharmaceutically acceptable salt thereof.
  • the present invention provides methods for treating, stabilizing or lessening the severity or progression of one or more diseases or disorders associated with one or both of ERK1 and ERK2, comprising administering to a patient in need thereof a composition comprising Compound 1, or a pharmaceutically acceptable salt thereof, and a TOR inhibitor, wherein Compound 1 is administered in unit dosage formulations that comprise between about about 5 mg to about 1000 mg of Compound 1.
  • a unit dosage formulation of the present invention provides about 1 mg, 5 mg, about 10 mg, about 15 mg, about 20 mg, about 25 mg, about 30 mg, about 35 mg, about 40 mg, about 45 mg, about 50 mg, about 55 mg, about 60 mg, about 65 mg, about 70 mg, about 75 mg, about 80 mg, about 85 mg, about 90 mg, about 95 mg, about 100 mg, about 105 mg, about 110 mg, about 115 mg, about 120 mg, about 125 mg, about 130 mg, about 135 mg, about 140 mg, about 145 mg, about 150 mg, about 155 mg, about 160 mg, about 165 mg, about 170 mg, about 175 mg, about 180 mg, about 185 mg, about 190 mg, about 195 mg, about 200 mg, about 225 mg, about 250 mg, about 275 mg, about 300 mg, about 325 mg, about 350 mg, about 375 mg, about 400 mg, about 425 mg, about 450 mg, about 475 mg, about 500 mg, about 550 mg, about 600 mg, about
  • the present invention provides methods for treating, stabilizing or lessening the severity or progression of one or more diseases or disorders associated with one or both of ERK1 and ERK2, comprising administering to a patient in need thereof a composition comprising Compound 1, or a pharmaceutically acceptable salt thereof, and a TOR inhibitor, wherein Compound 1 is administered in unit dosage formulations that comprise about 5 mg, 30 mg, or 150 mg of Compound 1.
  • a capsule formulation of the present invention provides about 25 mg, about 50 mg, about 75 mg, about 100 mg, about 125 mg, or about 150 mg of Compound 1.
  • Compound 1, or a pharmaceutically acceptable salt thereof is administered at dosage levels of about 0.01 mg/kg to about 50 mg/kg and preferably from about 1 mg/kg to about 25 mg/kg, of subject body weight per day, one or more times a day, to obtain the desired therapeutic effect.
  • Unit Dosage Forms of a TOR Inhibitor is administered at dosage levels of about 0.01 mg/kg to about 50 mg/kg and preferably from about 1 mg/kg to about 25 mg/kg, of subject body weight per day, one or more times a day, to obtain the desired therapeutic effect.
  • the present invention provides methods for treating, stabilizing or lessening the severity or progression of one or more diseases or disorders associated with one or both of ERK1 and ERK2, comprising administering to a patient in need thereof a composition comprising Compound 1, or a pharmaceutically acceptable salt thereof, and a TOR inhibitor, wherein the TOR inhibitor is administered in unit dosage formulations that comprise between about 0.1 mg and about 2000 mg, about 1 mg and 200 mg, about 35 mg and about 1400 mg, about 125 mg and about 1000 mg, about 250 mg and about 1000 mg, or about 500 mg and about 1000 mg of a TOR inhibitor.
  • unit dosage formulations comprising about 0.1 mg, 0.25 mg, 0.5 mg, 1 mg, 5 mg, 10 mg, 15 mg, 20 mg, 30 mg, 45 mg, 50 mg, 60 mg, 75 mg, 100 mg, 125 mg, 150 mg, 200 mg, 250 mg, 300 mg, 400 mg, 600 mg or 800 mg of a TOR inhibitor.
  • unit dosage formulations that comprise 0.1 mg, 0.25 mg, 0.5 mg, 1 mg, 2.5 mg, 5 mg, 10 mg, 15 mg, 20 mg, 30 mg, 35 mg, 50 mg, 70 mg, 100 mg, 125 mg, 140 mg, 175 mg, 200 mg, 250 mg, 280 mg, 350 mg, 500 mg, 560 mg, 700 mg, 750 mg, 1000 mg or 1400 mg of a TOR inhibitor.
  • unit dosage formulations that comprise about 5 mg, about 15 mg, about 20 mg, about 30 mg, about 45 mg, and about 50 mg of a TOR inhibitor.
  • unit dosage formulations that comprise about 2 mg, about 5 mg, about 7.5 mg, about 10 mg and about 15 mg of a TOR inhibitor.
  • Administration of Compound 1 is provided herein.
  • Compound 1, or a pharmaceutically acceptable salt thereof, and compositions thereof according to methods of the present invention are administered using any amount and any route of administration effective for treating or lessening the severity of a disorder provided above.
  • the exact amount required will vary from subject to subject, depending on the species, age, and general condition of the subject, the severity of the infection, the particular agent, its mode of administration, and the like.
  • provided methods comprise administering a pharmaceutically acceptable composition comprising Compound 1, or a pharmaceutically acceptable salt thereof, one, two, three, or four times a day.
  • a pharmaceutically acceptable composition comprising Compound 1, or a pharmaceutically acceptable salt thereof is administered once daily (“QD”).
  • QD once daily
  • a pharmaceutically acceptable composition comprising Compound 1, or a pharmaceutically acceptable salt thereof is administered twice daily.
  • twice daily administration refers to a compound or composition that is administered“BID”, or two equivalent doses administered at two different times in one day.
  • a pharmaceutically acceptable composition comprising Compound 1, or a pharmaceutically acceptable salt thereof, is administered three times a day.
  • a pharmaceutically acceptable composition comprising Compound 1, or a pharmaceutically acceptable salt thereof is administered“TID”, or three equivalent doses administered at three different times in one day.
  • a pharmaceutically acceptable composition comprising Compound 1, or a pharmaceutically acceptable salt thereof is administered four times a day.
  • a pharmaceutically acceptable composition comprising Compound 1, or a pharmaceutically acceptable salt thereof is administered“QID”, or four equivalent doses administered at four different times in one day.
  • Compound 1 is administered to a patient under fasted conditions and the total daily dose is any of those contemplated above and herein.
  • Compound 1 is administered to a patient under fed conditions and the total daily dose is any of those contemplated above and herein.
  • Compound 1 is administered orally.
  • compositions of this invention can be administered to humans and other animals orally, rectally, parenterally, intracisternally, intravaginally, intraperitoneally, topically (as by powders, ointments, or drops), bucally, as an oral or nasal spray, or the like, depending on the severity of the disease or disorder being treated.
  • Administration of a TOR Inhibitor a TOR Inhibitor
  • provided methods comprise administering a pharmaceutically acceptable composition comprising a TOR inhibitor one, two, three, or four times a day.
  • a pharmaceutically acceptable composition comprising a TOR inhibitor is administered once daily (“QD”).
  • QD once daily
  • a pharmaceutically acceptable composition comprising a TOR inhibitor is administered twice daily.
  • twice daily administration refers to a compound or composition that is administered“BID”, or two equivalent doses administered at two different times in one day.
  • a pharmaceutically acceptable composition comprising a TOR inhibitor is administered three times a day. In some embodiments, a pharmaceutically acceptable composition comprising a TOR inhibitor is administered“TID”, or three equivalent doses administered at three different times in one day.
  • a pharmaceutically acceptable composition comprising a TOR inhibitor is administered four times a day.
  • a pharmaceutically acceptable composition comprising a TOR inhibitor is administered“QID”, or four equivalent doses administered at four different times in one day.
  • a TOR inhibitor is administered to a patient under fasted conditions and the total daily dose is any of those contemplated above and herein.
  • a TOR inhibitor is administered to a patient under fed conditions and the total daily dose is any of those contemplated above and herein.
  • a TOR inhibitor is administered orally for reasons of convenience.
  • a TOR inhibitor when administered orally, is administered with a meal and water.
  • the TOR inhibitor is dispersed in water or juice (e.g., apple juice or orange juice) and administered orally as a suspension.
  • a TOR inhibitor when administered orally, is administered in a fasted state.
  • a TOR inhibitor can also be administered intradermally, intramuscularly, intraperitoneally, percutaneously, intravenously, subcutaneously, intranasally, epidurally, sublingually, intracerebrally, intravaginally, transdermally, rectally, mucosally, by inhalation, or topically to the ears, nose, eyes, or skin.
  • the mode of administration is left to the discretion of the health-care practitioner, and can depend in-part upon the site of the medical condition.
  • the present invention provides a pharmaceutically acceptable composition comprising Compound 1, or a pharmaceutically acceptable salt thereof. In some embodiments, the present invention provides a pharmaceutically acceptable composition of a TOR inhibitor. In some embodiments, a composition comprising Compound 1, or a pharmaceutically acceptable salt thereof, is separate from a composition comprising a TOR inhibitor. In some embodiments, Compound 1, or a pharmaceutically acceptable salt thereof, and a TOR inhibitor are present in the same composition.
  • Liquid dosage forms for oral administration include, but are not limited to, pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups and elixirs.
  • the liquid dosage forms may contain inert diluents commonly used in the art such as, for example, water or other solvents, solubilizing agents and emulsifiers such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, dimethylformamide, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor, and sesame oils), glycerol, tetrahydrofurfuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof.
  • inert diluents commonly used in the art such as, for example, water or other solvents,
  • Injectable preparations for example, sterile injectable aqueous or oleaginous suspensions may be formulated according to the known art using suitable dispersing or wetting agents and suspending agents.
  • the sterile injectable preparation may also be a sterile injectable solution, suspension or emulsion in a nontoxic parenterally acceptable diluent or solvent, for example, as a solution in 1,3-butanediol.
  • the acceptable vehicles and solvents that may be employed are water, Ringer's solution, U.S.P. and isotonic sodium chloride solution.
  • sterile, fixed oils are conventionally employed as a solvent or suspending medium.
  • any bland fixed oil can be employed including synthetic mono- or diglycerides.
  • fatty acids such as oleic acid are used in the preparation of injectables.
  • Injectable formulations can be sterilized, for example, by filtration through a bacterial-retaining filter, or by incorporating sterilizing agents in the form of sterile solid compositions which can be dissolved or dispersed in sterile water or other sterile injectable medium prior to use.
  • Injectable depot forms are made by forming microencapsule matrices of Compound 1, or a pharmaceutically acceptable salt thereof, and/or a TOR inhibitor, in biodegradable polymers such as polylactide-polyglycolide. Depending upon the ratio of compound to polymer and the nature of the particular polymer employed, the rate of compound release can be controlled. Examples of other biodegradable polymers include poly(orthoesters) and poly(anhydrides). Depot injectable formulations are also prepared by entrapping the compound in liposomes or microemulsions that are compatible with body tissues.
  • compositions for rectal or vaginal administration are preferably suppositories which can be prepared by mixing the compounds of this invention with suitable non-irritating excipients or carriers such as cocoa butter, polyethylene glycol or a suppository wax which are solid at ambient temperature but liquid at body temperature and therefore melt in the rectum or vaginal cavity and release the active compound.
  • suitable non-irritating excipients or carriers such as cocoa butter, polyethylene glycol or a suppository wax which are solid at ambient temperature but liquid at body temperature and therefore melt in the rectum or vaginal cavity and release the active compound.
  • Solid dosage forms for oral administration include capsules, tablets, pills, powders, and granules.
  • the active compound is mixed with at least one inert, pharmaceutically acceptable excipient or carrier such as sodium citrate or dicalcium phosphate and/or a) fillers or extenders such as starches, lactose, sucrose, glucose, mannitol, and silicic acid, b) binders such as, for example, carboxymethylcellulose, alginates, gelatin, polyvinylpyrrolidinone, sucrose, and acacia, c) humectants such as glycerol, d) disintegrating agents such as agar--agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and sodium carbonate, e) solution retarding agents such as paraffin, f) absorption accelerators such as quaternary ammonium compounds, g) wetting agents such as, for example, cety
  • Solid compositions of a similar type may also be employed as fillers in soft and hard- filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polyethylene glycols and the like.
  • the solid dosage forms of tablets, dragees, capsules, pills, and granules can be prepared with coatings and shells such as enteric coatings and other coatings well known in the pharmaceutical formulating art. They may optionally contain opacifying agents and can also be of a composition that they release the active ingredient(s) only, or preferentially, in a certain part of the intestinal tract, optionally, in a delayed manner. Examples of embedding compositions that can be used include polymeric substances and waxes. Solid compositions of a similar type may also be employed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polethylene glycols and the like.
  • Compound 1, or a pharmaceutically acceptable salt thereof, and/or a TOR inhibitor can also be in micro-encapsulated form with one or more excipients as noted above.
  • the solid dosage forms of tablets, dragees, capsules, pills, and granules can be prepared with coatings and shells such as enteric coatings, release controlling coatings and other coatings well known in the pharmaceutical formulating art.
  • Compound 1, or a pharmaceutically acceptable salt thereof, and/or a TOR inhibitor may be admixed with at least one inert diluent such as sucrose, lactose or starch.
  • Such dosage forms may also comprise, as is normal practice, additional substances other than inert diluents, e.g., tableting lubricants and other tableting aids such a magnesium stearate and microcrystalline cellulose.
  • additional substances other than inert diluents e.g., tableting lubricants and other tableting aids such a magnesium stearate and microcrystalline cellulose.
  • the dosage forms may also comprise buffering agents. They may optionally contain opacifying agents and can also be of a composition that they release the active ingredient(s) only, or preferentially, in a certain part of the intestinal tract, optionally, in a delayed manner. Examples of embedding compositions that can be used include polymeric substances and waxes.
  • Dosage forms for topical or transdermal administration of Compound 1, or a pharmaceutically acceptable salt thereof, and/or a TOR inhibitor include ointments, pastes, creams, lotions, gels, powders, solutions, sprays, inhalants or patches.
  • the active component is admixed under sterile conditions with a pharmaceutically acceptable carrier and any needed preservatives or buffers as may be required.
  • Ophthalmic formulation, ear drops, and eye drops are also contemplated as being within the scope of this invention.
  • the present invention contemplates the use of transdermal patches, which have the added advantage of providing controlled delivery of a compound to the body.
  • Such dosage forms can be made by dissolving or dispensing the compound in the proper medium.
  • Absorption enhancers can also be used to increase the flux of the compound across the skin. The rate can be controlled by either providing a rate controlling membrane or by dispersing the compound in a polymer matrix or gel.
  • the invention relates to a method of inhibiting protein kinase activity in a biological sample comprising the step of contacting said biological sample with Compound 1, or a pharmaceutically acceptable salt thereof, and/or a TOR inhibitor, or a composition comprising said compound.
  • the invention relates to a method of inhibiting one or both of ERK 1 and ERK2 kinase, or a mutant thereof, activity in a biological sample comprising the step of contacting said biological sample with Compound 1, or a pharmaceutically acceptable salt thereof, in combination with a TOR inhibitor, or a composition comprising any of the foregoing.
  • the invention relates to a method of irreversibly inhibiting one or both of ERK1 and ERK2 kinase, or a mutant thereof, activity in a biological sample comprising the step of contacting said biological sample with Compound 1, or a pharmaceutically acceptable salt thereof, in combination with a TOR inhibitor, or a composition comprising any of the foregoing.
  • biological sample includes, without limitation, cell cultures or extracts thereof; biopsied material obtained from a mammal or extracts thereof; and blood, saliva, urine, feces, semen, tears, or other body fluids or extracts thereof.
  • Inhibition of one or both of ERK1 and ERK2, or a mutant thereof, activity in a biological sample is useful for a variety of purposes that are known to one of skill in the art. Examples of such purposes include, but are not limited to, blood transfusion, organ- transplantation, biological specimen storage, and biological assays.
  • Another embodiment of the present invention relates to a method of inhibiting protein kinase activity in a patient comprising the step of administering to said patient Compound 1, or a pharmaceutically acceptable salt thereof, in combination with a TOR inhibitor, or a composition comprising any of the foregoing.
  • the invention relates to a method of irreversibly inhibiting one or both of ERK1 and ERK2 kinase, or a mutant thereof, activity in a patient comprising the step of administering to said patient Compound 1, or a pharmaceutically acceptable salt thereof, in combination with a TOR inhibitor, or a composition comprising any of the foregoing.
  • the activity is inhibited irreversibly by covalently modifying Cys 183 of ERK1. In certain embodiments, the activity is inhibited irreversibly by covalently modifying Cys 166 of ERK2. In certain embodiments, the activity is inhibited irreversibly by covalently modifying Cys 183 of ERK1 and Cys 166 of ERK2.
  • Pharmaceutically Acceptable Compositions of TOR Inhibitors are provided.
  • compositions comprising an effective amount of a TOR inhibitor and compositions comprising an effective amount of a TOR inhibitor and a pharmaceutically acceptable carrier or vehicle.
  • the pharmaceutical compositions described herein are suitable for oral, parenteral, mucosal, transdermal or topical administration.
  • TOR inhibitors can be administered to a patient orally or parenterally in the conventional form of preparations, such as capsules, microcapsules, tablets, granules, powder, troches, pills, suppositories, injections, suspensions and syrups.
  • Suitable formulations can be prepared by methods commonly employed using conventional, organic or inorganic additives, such as an excipient (e.g., sucrose, starch, mannitol, sorbitol, lactose, glucose, cellulose, talc, calcium phosphate or calcium carbonate), a binder (e.g., cellulose, methylcellulose, hydroxymethylcellulose, polypropylpyrrolidone, polyvinylpyrrolidone, gelatin, gum arabic, polyethyleneglycol, sucrose or starch), a disintegrator (e.g., starch, carboxymethylcellulose, hydroxypropylstarch, low substituted hydroxypropylcellulose, sodium bicarbonate, calcium phosphate or calcium citrate), a lubricant (e.g., magnesium stearate, light anhydrous silicic acid, talc or sodium lauryl sulfate), a flavoring agent (e.g., citric acid, menthol, glycine or orange powder
  • An effective amount of a TOR inhibitor in a pharmaceutical composition may be at a level that will exercise the desired effect; for example, about 0.005 mg/kg of a patient’s body weight to about 10 mg/kg of a patient’s body weight in unit dosage for both oral and parenteral administration.
  • capsules containing a TOR inhibitor without an additional carrier, excipient or vehicle.
  • compositions comprising an effective amount of a TOR inhibitor and a pharmaceutically acceptable carrier or vehicle, wherein a pharmaceutically acceptable carrier or vehicle can comprise an excipient, diluent, or a mixture thereof.
  • a pharmaceutically acceptable carrier or vehicle can comprise an excipient, diluent, or a mixture thereof.
  • the composition is a pharmaceutical composition.
  • compositions can be in the form of tablets, chewable tablets, capsules, solutions, parenteral solutions, troches, suppositories and suspensions and the like.
  • Compositions can be formulated to contain a daily dose, or a convenient fraction of a daily dose, in a dosage unit, which may be a single tablet or capsule or convenient volume of a liquid.
  • the solutions are prepared from water-soluble salts, such as the hydrochloride salt.
  • all of the compositions are prepared according to known methods in pharmaceutical chemistry.
  • Capsules can be prepared by mixing a TOR inhibitor with a suitable carrier or diluent and filling the proper amount of the mixture in capsules.
  • the usual carriers and diluents include, but are not limited to, inert powdered substances such as starch of many different kinds, powdered cellulose, especially crystalline and microcrystalline cellulose, sugars such as fructose, mannitol and sucrose, grain flours and similar edible powders.
  • Tablets can be prepared by direct compression, by wet granulation, or by dry granulation. Their formulations usually incorporate diluents, binders, lubricants and disintegrators as well as the compound. Typical diluents include, for example, various types of starch, lactose, mannitol, kaolin, calcium phosphate or sulfate, inorganic salts such as sodium chloride and powdered sugar. Powdered cellulose derivatives are also useful. In some embodiments, the pharmaceutical composition is lactose-free. Typical tablet binders are substances such as starch, gelatin and sugars such as lactose, fructose, glucose and the like. Natural and synthetic gums are also convenient, including acacia, alginates, methylcellulose, polyvinylpyrrolidine and the like. Polyethylene glycol, ethylcellulose and waxes can also serve as binders.
  • a lubricant might be necessary in a tablet formulation to prevent the tablet and punches from sticking in the die.
  • the lubricant can be chosen from such slippery solids as talc, magnesium and calcium stearate, stearic acid and hydrogenated vegetable oils.
  • Tablet disintegrators are substances that swell when wetted to break up the tablet and release the compound. They include starches, clays, celluloses, algins and gums. More particularly, corn and potato starches, methylcellulose, agar, bentonite, wood cellulose, powdered natural sponge, cation-exchange resins, alginic acid, guar gum, citrus pulp and carboxymethyl cellulose, for example, can be used as well as sodium lauryl sulfate. Tablets can be coated with sugar as a flavor and sealant, or with film-forming protecting agents to modify the dissolution properties of the tablet.
  • the compositions can also be formulated as chewable tablets, for example, by using substances such as mannitol in the formulation.
  • Cocoa butter is a traditional suppository base, which can be modified by addition of waxes to raise its melting point slightly.
  • Water-miscible suppository bases comprising, particularly, polyethylene glycols of various molecular weights are in wide use.
  • the effect of the TOR inhibitor can be delayed or prolonged by proper formulation.
  • a slowly soluble pellet of the TOR inhibitor can be prepared and incorporated in a tablet or capsule, or as a slow-release implantable device.
  • the technique also includes making pellets of several different dissolution rates and filling capsules with a mixture of the pellets. Tablets or capsules can be coated with a film that resists dissolution for a predictable period of time. Even the parenteral preparations can be made long-acting, by dissolving or suspending the TOR inhibitor in oily or emulsified vehicles that allow it to disperse slowly in the serum.
  • Compound 1 is prepared according to the following general procedure.
  • Step 2 can be carried out by adding Intermediate 1 to a suitable coupling partner in the presence of Na 2 CO 3 , a degassed solvent (e.g., tert-amyl alcohol), a suitable palladium catalyst (e.g., tris-dibenzylamino dipalladium) and a suitable phosphine ligand (e.g., Dave Phos) under conditions suitable to effect coupling.
  • a degassed solvent e.g., tert-amyl alcohol
  • a suitable palladium catalyst e.g., tris-dibenzylamino dipalladium
  • a suitable phosphine ligand e.g., Dave Phos
  • This Example describes experiments relating to the effect of the combination of Compound 1 (free base) with Compound 2 on the Panc1 (a.k.a. PANC-1) pancreatic cancer cell line, the Mia PaCa pancreatic cancer cell line, the HS294T melanoma cell line, the HCT-116 colorectal cancer cell line, the NCI-H460 lung cancer cell line, the NCI-H522 lung cancer cell line, or the NCI-H1755 lung cancer cell line.
  • cells were plated at a density of 3000 cells/well in 90 ⁇ L of growth media on 96 well clear bottom black-well plates (Corning Cat# 3904) and incubated overnight under standard cell culture growth conditions at 37 0 C 5% CO 2 .
  • the outer most rows and columns of wells were filled with culture media, without cells, to avoid evaporation effects on subsequent readouts.
  • the final concentrations in treatment wells for each compound was: (in nM) 10000.00, 3333.33, 1111.11, 370.37, 123.46, 41.15, 13.72, 4.57, 1.52, 0.
  • Each treatment was contained in 10 ⁇ L treatment media added to the respective wells.
  • Each concentration was tested in triplicate.
  • Panc1 (a.k.a., PANC-1) Pancreatic Cancer Cell Line: Panc 1 cells were treated with a 9-point dose dilution of Compound 1 or Compound 2 or both, ranging from 1.52 nM to 10000.00 nM and DMSO vehicle control.
  • Fig. 1A dose response to Compound 2 (trend line with triangles);
  • Fig. 1B dose response to Compound 1 (trend line with circles);
  • Fig. 1C combination treatment with Compound 1 and Compound 2 (trend line with circles).
  • Compound 2 induces cytostatic (growth-inhibitory) effect but not cell death, as noted by the dose-response curve remaining above the X-axis.
  • Compound 1 induces cell-death (cytotoxic) effect (noted by the curve crossing the X-axis) at 1111.11 nM, and the combination of Compound 1 and Compound 2 induces a cell death effect of the same magnitude as Compound 1 (since the curves plateau at the same level below x-axis between -30.1% and - 34.5% of starting cell growth control) but at a lower dose (370.37 nM of each agent).
  • Effects of Compound 1 and Compound 2 Used Alone or in Combination with Each Other on Viability of Mia PaCa Pancreatic Cancer Cell Line ( Figure 2).
  • FIG. 2A dose response to Compound 2
  • Fig. 2B dose response to Compound 1 (trend line with circles)
  • Fig. 2C combination treatment with Compound 1 and Compound 2 (trend line with circles).
  • Compound 2 induces cytostatic (growth-inhibitory) effect but not cell death, as noted by the curve remaining above the X-axis.
  • Compound 1 induces cell death (noted by the curve crossing the X-axis) at 247 nM
  • the combination of Compound 1 and Compound 2 induces a cell death effect of increased magnitude (since the curve of the Compound 1 and Compound 2 combination response plateaus at the -84.6% of starting growth control compared to -49.8% induced by Compound 1 alone) at a dose of 123.46 nM of each agent combined.
  • Effects of Compound 1 and Compound 2 Used Alone or in Combination with Each Other on Viability of HS294T Melanoma Cell Line Figure 3).
  • Figure 3. HS294T were treated with a 9-point dose dilution of Compound 1, Compound 2, or both, ranging from 1.52 nM to 10000.00 nM and DMSO vehicle control.
  • Fig. 3A dose response to Compound 2 (trend line with triangles);
  • Fig. 3B dose response to Compound 1 (trend line with circles);
  • Fig. 3C combination treatment with Compound 1 and Compound 2 (trend line with circles).
  • FIG. 4A HCT-116 cells were treated with a 9-point dose dilution of Compound 1 (Fig. 4A) or Compound 2 (Fig. 4B) ranging from 1.52 nM to 10000.00 nM and DMSO vehicle control. Combination treatment with Compound 1 and Compound 2 (Fig. 4C).
  • Compound 2 induces cytostatic (growth-inhibitory) effect but not cell death, as noted by the dose-response curve remaining above the X-axis.
  • Compound 1 induces cell-death (cytotoxic) effect (noted by the curve crossing the X-axis) at 1321.30 nM.
  • Combination of Compound 1 and Compound 2 induces cell death effect at a lower dose (243.78 nM) than treatment with Compound 1 alone and of larger magnitude (88.3 compared to 65.3 percent cell death induced by Compound 1 alone). Effects of Compound 1 and Compound 2 Used Alone or in Combination with Each Other on Viability of NCI-H460 Lung Cancer Cell Line ( Figure 5).
  • FIG. 5A NCI-H460 cells were treated with a 9-point dose dilution of Compound 1 (Fig. 5A) or Compound 2 (Fig. 5B) ranging from 1.52 nM to 10000.00 nM and DMSO vehicle control. Combination treatment with Compound 1 and Compound 2 (Fig. 5C).
  • Compound 2 induces cytostatic (growth-inhibitory) effect but not cell death, as noted by the dose-response curve remaining above the X-axis.
  • Compound 1 induces cell-death (cytotoxic) effect (noted by the curve crossing the X-axis) at 6839.12 nM.
  • Combination of Compound 1 and Compound 2 induces cell death effect at a lower dose (1857.8 nM) than treatment with Compound 1 alone and of larger magnitude (66.3 compared to 18.0 percent cell death induced by Compound 1 alone). Effects of Compound 1 and Compound 2 Used Alone or in Combination with Each Other on Viability of NCI-H522 Lung Cancer Cell Line ( Figure 6).
  • FIG. 6A NCI-H522 cells were treated with a 9-point dose dilution of Compound 1 (Fig. 6A) or Compound 2 (Fig. 6B) ranging from 1.52 nM to 10000.00 nM and DMSO vehicle control. Combination treatment with Compound 1 and Compound 2 (Fig. 6C).
  • Compound 2 induces cytostatic (growth-inhibitory) effect but not cell death, as noted by the dose-response curve remaining above the X-axis.
  • Compound 1 induces cell-death (cytotoxic) effect (noted by the curve crossing the X-axis) at 2338.84 nM.
  • Combination of Compound 1 and Compound 2 induces cell death effect at a lower dose (625.17 nM) than treatment with Compound 1 alone and of larger magnitude (88.2 compared to 55.7 percent cell death induced by Compound 1 alone ). Effects of Compound 1 and Compound 2 Used Alone or in Combination with Each Other on Viability of NCI-H1755 Lung Cancer Cell Line ( Figure 7).
  • FIG. 7 NCI-H1755 cells were treated with a 9-point dose dilution of Compound 1 (Fig. 7A) or Compound 2 (Fig. 7B) ranging from 1.52 nM to 10000.00 nM and DMSO vehicle control. Combination treatment with Compound 1 and Compound 2 (Fig. 7C).
  • Compound 2 induces cytostatic (growth-inhibitory) effect but not cell death, as noted by the dose-response curve remaining above the X-axis.
  • Compound 1 induces cell-death (cytotoxic) effect (noted by the curve crossing the X-axis) at 722.77 nM.
  • Combination of Compound 1 and Compound 2 induces cell death effect at a lower dose (295.12 nM) than treatment with Compound 1 alone (370.37 nM). Effects of Compound 1 and Compound 2 Used Alone or in Combination with Each Other on Viability of Mia PaCa Pancreatic Cancer Cell Line ( Figure 21).
  • FIG. 21 This experiment is a replicate of the above-described assay on Mia PaCa cells.
  • Figure 21 Mia PaCa Pancreatic Cancer Cell Line: Mia PaCa cells were treated with a 9-point dose dilution of Compound 1, or Compound 2, or both, ranging from 1.52 nM to 10000.00 nM, and DMSO vehicle control.
  • Fig. 21A dose response to Compound 1 alone (trend line with circles) or Compound 2 alone (trend line with triangles);
  • Fig. 21B combination treatment with Compound 1 and Compound 2 (trend line with circles) and untreated (trendline with triangles).
  • Compound 2 alone induces cytostatic (growth-inhibitory) effect but not cell death, as noted by the dose-response curve remaining above the X-axis.
  • Compound 1 alone induces cell-death (cytotoxic) effect (noted by the curve crossing the X-axis) at 1238.80 nM.
  • Combination of Compound 1 and Compound 2 induces cell death effect at a lower dose (533.33 nM) than treatment with Compound 1 alone and of larger magnitude (92.1 compared to 66.5 percent cell death induced by Compound 1 alone ). Effects of Compound 1 and Compound 2 Used Alone or in Combination with Each Other on Viability of Mia PaCa Pancreatic Cancer Cell Line ( Figure 22).
  • FIG. 22 This experiment is a replicate of the above-described assay on Mia PaCa cells.
  • Figure 22. Mia PaCa Pancreatic Cancer Cell Line: Mia PaCa cells were treated with a 9-point dose dilution of Compound 1, or Compound 2, or both, ranging from 1.52 nM to 10000.00 nM, and DMSO vehicle control.
  • Fig. 22A dose response to Compound 1 alone (trend line with circles) or Compound 2 alone (trend line with triangles);
  • Fig. 22B combination treatment with Compound 1 and Compound 2 (trend line with circles).
  • Compound 2 induces cytostatic (growth-inhibitory) effect but not cell death, as noted by the dose-response curve remaining above the X-axis.
  • Compound 1 induces cell-death (cytotoxic) effect (noted by the curve crossing the X-axis) at 1534.61 nM.
  • Combination of Compound 1 and Compound 2 induces cell death effect at a lower dose (628.06 nM) than treatment with Compound 1 alone and of larger magnitude (88.9 compared to 49.9 percent cell death induced by Compound 1 alone ). Effects of Compound 1 and Compound 2 Used Alone or in Combination with Each Other on Viability of HS294T Melanoma Cancer Cell Line ( Figure 23).
  • FIG. 23 HS294T MelanomaCancer Cell Line: HS294T cells were treated with a 9-point dose dilution of Compound 1, or Compound 2, or both, ranging from 1.52 nM to 10000.00 nM, and DMSO vehicle control.
  • Fig. 23A dose response to Compound 1 alone (trend line with circles) or Compound 2 alone (trend line with triangles);
  • Fig. 23B combination treatment with Compound 1 and Compound 2 (trend line with circles).
  • Compound 2 alone induces cytostatic (growth-inhibitory) effect but not cell death, as noted by the dose-response curve remaining above the X-axis.
  • Compound 1 alone induces cell-death (cytotoxic) effect (noted by the curve crossing the X-axis) at 1202.26 nM.
  • Combination of Compound 1 and Compound 2 induces cell death effect at a lower dose (276.0 nM) than treatment with Compound 1 alone and of larger magnitude (95.6 compared to 86.9 percent cell death induced by Compound 1 alone).
  • Emax is the effect a compound has on cell growth at a given concentration It is evaluated both relative to control treatment and relative to cell number at time zero, and usually ranges from 100% to - 100%.
  • an Emax of 100% indicates that the treated cells have grown as fast as the control cells; an Emax of 20% indicates that the treated cells have grown slower and their growth rate was 20% of the growth rate of cells in control treated wells; an Emax of -20% indicates that the cells have declined in number compared to the number of cells seeded at day zero (i.e., by 20%); an Emax of -100% indicates that the cells have declined in number compared to the number of cells seeded at day zero (i.e., by 100%, that is, all cells are dead). Values of >100% indicate that the cells in the treated well have grown faster than the cells in the control well, typically indicating no negative effect (as opposed to a positive effect).
  • Calcusyn Analysis The following assays were conducted with Compound 1 and Compound 2 and demonstrate that in certain cell lines synergy is observed upon treatment with a combination of Compound 1 and Compound 2. The Calcusyn assay was carried out over 5 days. On day 1, cells were plated into 96 well plates, in 100 ⁇ L media (cell number calculated to give 50-70% fluency on day 5). On day 2, drug dilutions were prepared at 2x final concentration in media, 100 L /well were added, and 4-5 replicates/drug concentration were completed. On day 5, media was removed from the plate. 100 ⁇ L were added of 10% TCA/well and fixed for 1 hour at 4 °C.
  • B-raf mutant vemurafenib-resistant melanoma cell line 1 (BRAF status: V600E(homo)); B-raf mutant vemurafenib-resistant melanoma cell line 2 (BRAF status: V600E(het)); B-raf mutant vemurafenib-resistant melanoma cell line 3 (BRAF status: V600E(homo)); B-raf mutant vemurafenib-resistant melanoma cell line 4 (BRAF status: V600E(het)); B-raf mutant vemurafenib-resistant melanoma cell 5 (BRAF status: V600 E(het)); B-raf mutant vemurafenib-resistant melanoma cell 5 (BRAF status: V600 E(het)); B-raf mutant vemurafenib-resistant melanoma cell 5 (BRAF status: V600 E(het
  • Colony assays were performed to determine the ability of a B-raf mutant vemurafenib-resistant melanoma cell line (i.e., cell line 8) to generate colonies after treatment with a combination of Compounds 1 and 2.
  • Figure 10 contains a representative graph depicting the results of these assays.
  • Colony assays were performed to determine the ability of a B-raf mutant vemurafenib-resistant melanoma cell line (i.e., cell line 3) to generate colonies after treatment with a combination of Compounds 1 and 2.
  • Figure 11 contains a representative graph depicting the results of these assays.
  • a combination PD study of Compound 1 (in the form of the phosphate salt) and Compound 6 was run in a KRAS G12D cell line CRC (i.e., colorectal cancer) PDX model. Briefly, the study design is as described below. Greater than 60 athymic nude mice were implanted with KRAS G12D CRC patient-derived xenographs. The drugs were formulated in 5% Captisol and 0.4% Tween80. The mice were treated Monday through Friday for 28 days, or until tumors reached 1250 mm 3 . Plasma was isolated on day 28 at 0.5, 1, 2, 4, 8 and 24 hours. The tumors were harvested on day 28 with 1/3 of the tumor formalin fixed and the remaining 2/3 of the tumor snap frozen.
  • CRC colorectal cancer
  • the cells were plated at density 3000 cells/well in 90 ⁇ L of growth media on 96 well clear bottom black-well plates (Corning Cat# 3904) and incubated overnight under standard cell culture growth conditions at 37 0 C, 5% CO 2 .
  • the outer most rows and columns of wells were filled with culture media, without cells, to avoid evaporation effects on subsequent readouts.
  • the final concentrations of Compound 1 in treatment wells were: (in nM) 10000.00, 3333.33, 1111.11, 370.37, 123.46, 41.15, 13.72, 4.57, 1.52, and 0.
  • Compound 1 was combined at each dilution level with a selected constant dose of Compound 2.
  • Each treatment was contained in 10 ⁇ L treatment media added to the respective wells.
  • Each concentration was tested in triplicate.
  • FIG. 14-17, 19-20, and 24-29 depict volcano plots generated using data generated in the manner described above in order to detect synergy for combination drug treatment of certain cell lines. Assays were completed as described above using Compound 1 and Compound 2.
  • the x-axis shows the 9 point dose titration of Compound 1 and DMSO control; the y-axis shows three individual fixed concentrations of Compound 2 in each experiment; and the z-axis shows the magnitude of the effect, with different patterns denoting different percent improvement over the additive effect of the specific compound combinations, as specified in each figure.
  • synergy is defined as achieving a greater than additive effect with a particular drug combination.
  • An additive effect is a predicted additive effect, assuming independent action of each individual drug (i.e., Bliss independence). If a particular combination of drugs affords results which exceed the predicted additive effect, that combination is considered to be synergistic. If a particular combination of drugs affords results which fall short of the predicted additive effect, that combination is considered to be antagonistic.
  • Figure 14 depicts a volcano plot at section (a) generated using data obtained for HCT-116 colorectal cancer cells treated with a combination of Compound 1 and Compound 2 at concentrations of 0.1 M, 0.3 ⁇ M and 1.0 ⁇ M. The height of the plot peak suggests a synergistic effect is achieved at certain combination doses. Relevant dose response assay data are provided at section (b) of Figure 14.
  • Figure 15 depicts a volcano plot at section (a) generated using data obtained for HCT-116 colorectal cancer cells treated with a combination of Compound 1 and Compound 2 at concentrations of 0.03 ⁇ M, 0.3 ⁇ M and 1.0 ⁇ M. The height of the plot peak suggests a synergistic effect is achieved at certain combination doses. Relevant dose response assay data are provided at section (b) of Figure 15.
  • Figure 16 depicts a volcano plot at section (a) generated using data obtained from a replicate experiment for HCT-116 colorectal cancer cells treated with a combination of Compound 1 and Compound 2 at concentrations of 0.03 ⁇ M, 0.3 ⁇ M and 1.0 ⁇ M. The height of the plot peak suggests a synergistic effect is achieved at certain combination doses. Relevant dose response assay data are provided at section (b) of Figure 16.
  • Figure 17 depicts a volcano plot at section (a) generated using data obtained for MiaPaCa pancreatic cancer cells treated with a combination of Compound 1 and Compound 2 at concentrations of 0.1 ⁇ M, 0.3 ⁇ M and 1.0 ⁇ M. The height of the plot peak suggests an additive effect is achieved at certain combination doses. Relevant dose response assay data are provided at section (b) of Figure 17.
  • Figure 19 depicts a volcano plot at section (a) generated using data obtained for MiaPaCa pancreatic cancer cells treated with a combination of Compound 1 and Compound 2 at concentrations of 0.03 ⁇ M, 0.3 ⁇ M and 1.0 ⁇ M. The height of the plot peak suggests a an additive effect is achieved at certain combination doses. Relevant dose response assay data are provided at section (b) of Figure 19.
  • Figure 20 depicts a volcano plot at section (a) generated using data obtained from a replicate experiment for MiaPaCa pancreatic cancer cells treated with a combination of Compound 1 and Compound 2 at concentrations of 0.03 ⁇ M, 0.3 ⁇ M and 1.0 ⁇ M. The height of the plot peak suggests additive effect is achieved at certain combination doses. Relevant dose response assay data are provided at section (b) of Figure 20.
  • Figure 24 depicts a volcano plot generated using data obtained for Calu-1 lung cancer cells with a combination of Compound 1 and Compound 2 at concentrations of 0.1 ⁇ M, 0.3 ⁇ M and 1.0 ⁇ M. This plot suggests that additive effects are observed for this combination at certain dosages.

Abstract

The present invention provides methods of treating, stabilizing or lessening the severity or progression of a disease or disorder associated with one or both of ERK1 and ERK2.

Description

COMBINATIONS OF AN ERK INHIBITOR AND A TOR INHIBITOR AND RELATED
METHODS CROSS-REFERENCE TO RELATED CASES
[0001] The present application claims priority to U.S. provisional application number 62/037,067, filed August 13, 2014, the entirety of which is hereby incorporated by reference. FIELD OF THE INVENTION
[0002] The present invention provides methods of treating, stabilizing or lessening the severity or progression of a disease or disorder associated with one or both of ERK1 and ERK2 protein kinase. BACKGROUND OF THE INVENTION
[0003] The search for new therapeutic agents has been greatly aided in recent years by a better understanding of the structure of enzymes and other biomolecules associated with diseases. One important class of enzymes that has been the subject of extensive study is protein kinases.
[0004] Protein kinases constitute a large family of structurally related enzymes that are responsible for the control of a variety of signal transduction processes within the cell. Protein kinases are thought to have evolved from a common ancestral gene due to the conservation of their structure and catalytic function. Almost all kinases contain a similar 250-300 amino acid catalytic domain. The kinases may be categorized into families by the substrates they phosphorylate (e.g., protein-tyrosine, protein-serine/threonine, lipids, etc.).
[0005] The processes involved in tumor growth, progression, and metastasis are mediated by signaling pathways that are activated in cancer cells. The MAPK or Raf-Mek-ERK pathway plays a central role in regulating mammalian cell growth by relaying extracellular signals from ligand-bound cell surface tyrosine kinase receptors such as erbB family, PDGF, FGF, and VEGF receptor tyrosine kinase. Activation of the ERK occurs via a cascade of phosphorylation events that begins with activation of Ras. Activation of Ras leads to the recruitment and activation of Raf, a serine-threonine kinase. Activated Raf then phosphorylates and activates MEK1/2, which then phosphorylates and activates one or both of ERK1 and ERK2. When activated, one or both of ERK1 and ERK2 phosphorylates several downstream targets involved in a multitude of cellular events including cytoskeletal changes and transcriptional activation. The ERK/MAPK pathway is one of the most important for cell proliferation, and human tumor data suggest that the ERK/MAPK pathway is frequently activated in many tumors. Ras genes, which are upstream of one or both of ERK1 and ERK2, are mutated in several cancers including colorectal, melanoma, breast, lung, and pancreatic tumors. High Ras activity is accompanied by elevated ERK activity in many human tumors. In addition, activating mutations of BRAF, a serine- threonine kinase of the Raf family, are associated with increased RAF, MEK, and ERK kinase activity. Tumors types with the most frequent mutations in BRAF include melanomas (60%), thyroid cancers (greater than 40%) and colorectal cancers.
[0006] Many diseases are associated with abnormal cellular responses, proliferation and evasion of programmed cell-death, triggered by protein kinase-mediated events as described above. Accordingly, there remains a need to find protein kinase inhibitors useful as therapeutic agents. SUMMARY OF THE INVENTION
[0007] In some embodiments, the present invention provides methods of treating, stabilizing or lessening the severity or progression of one or more diseases or disorders associated with one or both of ERK1 and ERK2 comprising administering to a patient in need thereof an inhibitor of one or both of ERK 1 and ERK2 in combination with a TOR (i.e., target of rapamycin) inhibitor. In some aspects, the inhibitor of one or both of ERK1 and ERK2 is Compound 1 (N-(2-((2-((2- methoxy-5-methylpyridin-4-yl)amino)-5-(trifluoromethyl)pyrimidin-4-yl)amino)-5- methylphenyl)acrylamide):
Figure imgf000003_0001
or a pharmaceutically acceptable salt thereof.
[0008] Compound 1, N-(2-((2-((2-methoxy-5-methylpyridin-4-yl)amino)-5- (trifluoromethyl)pyrimidin-4-yl)amino)-5-methylphenyl)acrylamide, is designated as compound number I-90 in PCT patent application serial number PCT/US14/15256, filed February 7, 2014 and published as WO2014/124230 on August 14, 2014 (referred to herein as“the ‘230 publication,”) the entirety of which is hereby incorporated by reference. The synthesis of Compound 1 is described in detail at Example 94 of the‘230 publication. Compound 1 is active in a variety of assays and therapeutic models demonstrating covalent, irreversible inhibition of one or both of ERK1 and ERK2 kinases (see, e.g., Table A of the ‘230 publication). Accordingly, Compound 1, or a pharmaceutically acceptable salt thereof, is useful for treating one or more disorders associated with activity of one or both of ERK1 and ERK2, as described in detail herein, infra.
[0009] Additional embodiments describing methods of utilizing a provided combination are described in detail herein, infra. BRIEF DESCRIPTION OF THE DRAWINGS
[0010] Figures 1A, 1B and 1C. Panc1 (a.k.a., PANC-1) Pancreatic Cancer Cell Line: Panc 1 cells were treated with a 9-point dose dilution of Compound 1 or Compound 2 or both, ranging from 1.52 nM to 10000.00 nM and DMSO vehicle control. Fig. 1A: dose response to Compound 2 (trend line with triangles); Fig. 1B: dose response to Compound 1 (trend line with circles); Fig. 1C: combination treatment with Compound 1 and Compound 2 (trend line with circles).
[0011] Figures 2A, 2B and 2C. Mia PaCa Pancreatic Cancer Cell Line: Mia PaCa cells were treated with a 9-point dose dilution of Compound 1, or Compound 2, or both, ranging from 1.52 nM to 10000.00 nM, and DMSO vehicle control. Fig. 2A: dose response to Compound 2; Fig. 2B: dose response to Compound 1 (trend line with circles); Fig. 2C: combination treatment with Compound 1 and Compound 2 (trend line with circles).
[0012] Figures 3A, 3B and 3C. HS294T were treated with a 9-point dose dilution of Compound 1, Compound 2, or both, ranging from 1.52 nM to 10000.00 nM and DMSO vehicle control. Fig. 3A: dose response to Compound 2 (trend line with triangles); Fig. 3B: dose response to Compound 1 (trend line with circles); Fig. 3C: combination treatment with Compound 1 and Compound 2 (trend line with circles).
[0013] Figures 4A, 4B and 4C. HCT-116 cells were treated with a 9-point dose dilution of Compound 1 (Fig. 4A) or Compound 2 (Fig. 4B) ranging from 1.52 nM to 10000.00 nM and DMSO vehicle control. Combination treatment with Compound 1 and Compound 2 (Fig. 4C).
[0014] Figures 5A, 5B and 5C. NCI-H460 cells were treated with a 9-point dose dilution of Compound 1 (Fig. 5A) or Compound 2 (Fig. 5B) ranging from 1.52 nM to 10000.00 nM and DMSO vehicle control. Combination treatment with Compound 1 and Compound 2 (Fig. 5C).
[0015] Figures 6A, 6B and 6C. NCI-H522 cells were treated with a 9-point dose dilution of Compound 1 (Fig. 6A) or Compound 2 (Fig. 6B) ranging from 1.52 nM to 10000.00 nM and DMSO vehicle control. Combination treatment with Compound 1 and Compound 2 (Fig. 6C).
[0016] Figures 7A, 7B and 7C. NCI-H1755 cells were treated with a 9-point dose dilution of Compound 1 (Fig. 7A) or Compound 2 (Fig. 7B) ranging from 1.52 nM to 10000.00 nM and DMSO vehicle control. Combination treatment with Compound 1 and Compound 2 (Fig. 7C).
[0017] Figure 8. Emax values are shown against a panel of cancer cell lines. A moderate enhancement in Emax is seen when combining Compound 1 with Compound 2, a TOR inhibitor. Compound 1 does not kill any of the tested lines at 1 μM, and in combination with Compound 2, cell death is observed in 1/6 tested lines (i.e., NCI-H727). Emax in this line reached -16% when Compound 1 was combined with 1μM Compound 2
[0018] Figure 9. The results of a Calcusyn analysis to detect synergy of Compound 1 and Compound 2 combination treatement are shown for a panel of B-raf mutant vemurafenib resistant melanoma cell lines.
[0019] Figure 10. Activity in a BRAF-mutant vermurafenib-resistant melanoma cell line (i.e., cell line 8) colony forming assay is shown for the combination of Compound 1 and Compound 2.
[0020] Figure 11. Activity in a BRAF-mutant vemurafenib-resistant melanoma cell line (i.e., cell line 3) colony forming assay is shown for the combination of Compound 1 and Compound 2.
[0021] Figure 12. CI values are shown for the combination of Compound 1 and Compound 2 in NRAS mutant melanoma cell lines A-D. [0022] Figure 13. Depicts the results for Compound 1 (phosphate salt) and Compound 6 singly and in combination in the KRAS G12D cell line CRC PDX model.
[0023] Figure 14. Volcano plot at section (a) for HCT-116 colorectal cancer cells treated with a combination of Compound 1 (9-point dose dilution) and Compound 2 at concentrations of 0.1 μM, 0.3 μM and 1.0 μM . Relevant dose response assay data are provided at section (b).
[0024] Figure 15. Volcano plot at section (a) for HCT-116 colorectal cancer cells treated with a combination of Compound 1 (9-point dose dilution) and Compound 2 (at concentrations of 0.1 μM, 0.3 μM and 1.0 μM). Relevant dose response assay data are provided at section (b).
[0025] Figure 16. Volcano plot at section (a) for HCT-116 colorectal cancer cells treated with a combination of Compound 1 (9-point dose dilution) and Compound 2 (at concentrations of 0.1 μM, 0.3 μM and 1.0 μM). Relevant dose response assay data are provided at section (b).
[0026] Figure 17. Volcano plot at section (a) for Mia Paca pancreatic cancer cells treated with a combination of Compound 1 (9-point dose dilution) and Compound 2 (at concentrations of 0.1 μM, 0.3 μM and 1.0 μM). Relevant dose response assay data are provided at section (b).
[0027] Figure 18. Dose response assay data for Mia Paca pancreatic cancer cells treated with a combination of Compound 1 and Compound 2 (at concentrations of 0.1 μM, 0.3 μM and 1.0 μM).
[0028] Figure 19. Volcano plot at section (a) for Mia Paca pancreatic cancer cells treated with a combination of Compound 1 (9-point dose dilution) and Compound 2 (at concentrations of 0.03 μM, 0.3 μM and 1.0 μM) (run 1). Relevant dose response assay data are provided at section (b).
[0029] Figure 20. Volcano plot at section (a) for Mia Paca pancreatic cancer cells treated with a combination of Compound 1 (9-point dose dilution) and Compound 2 (at concentrations of 0.03 μM, 0.3 μM and 1.0 μM) (run 2). Relevant dose response assay data are provided at section (b).
[0030] Figure 21. Mia PaCa Pancreatic Cancer Cell Line: Mia PaCa cells were treated with a 9-point dose dilution of Compound 1, or Compound 2, or both, ranging from 1.52 nM to 10000.00 nM, and DMSO vehicle control. Fig. 21A: dose response to Compound 1 alone (trend line with circles) or Compound 2 alone (trend line with triangles); Fig. 21B: combination treatment with Compound 1 and Compound 2 (trend line with circles) and untreated (trendline with triangles).
[0031] Figure 22. Mia PaCa Pancreatic Cancer Cell Line: Mia PaCa cells were treated with a 9-point dose dilution of Compound 1, or Compound 2, or both, ranging from 1.52 nM to 10000.00 nM, and DMSO vehicle control. Fig. 22A: dose response to Compound 1 alone (trend line with circles) or Compound 2 alone (trend line with triangles); Fig. 22B: combination treatment with Compound 1 and Compound 2 (trend line with circles).
[0032] Figure 23. HS294T Melanoma Cancer Cell Line: HS294T cells were treated with a 9-point dose dilution of Compound 1, or Compound 2, or both, ranging from 1.52 nM to 10000.00 nM, and DMSO vehicle control. Fig. 23A: dose response to Compound 1 alone (trend line with circles) or Compound 2 alone (trend line with triangles); Fig. 23B: combination treatment with Compound 1 and Compound 2 (trend line with circles).
[0033] Figure 24. Volcano plot for Calu-1 lung cancer cells treated with a combination of Compound 1 (9-point dose dilution) and Compound 2 (at concentrations of 0.1 μM, 0.3 μM and 1.0 μM) (run 2).
[0034] Figures 25A and 25B. Volcano plot for HCT-116 colorectal cancer cells treated with a combination of Compound 1 (9-point dose dilution) and Compound 2 (at concentrations of 0.1 μM, 0.3 μM and 1.0 μM).
[0035] Figures 26Aand 26B. Volcano plot for Mia PaCa pancreatic cancer cells treated with a combination of Compound 1 (9-point dose dilution) and Compound 2 (at concentrations of 0.1 μM, 0.3 μM and 1.0 μM).
[0036] Figures 27A, 27B, 27C and 27D. Volcano plot for Calu-6 lung cancer cells treated with a combination of Compound 1 (9-point dose dilution) and Compound 2 (at concentrations of 0.1 μM, 0.3 μM and 1.0 μM).
[0037] Figures 28A and 28B. Volcano plot for NCI-H460 cells treated with a combination of Compound 1 (9-point dose dilution) and Compound 2 (at concentrations of 0.1 μM, 0.3 μM and 1.0 μM).
[0038] Figures 29A and 29B. Volcano plot for NCI-H727 cells treated with a combination of Compound 1 (9-point dose dilution) and Compound 2 (at concentrations of 0.1 μM, 0.3 μM and 1.0 μM).  
DETAILED DESCRIPTION OF THE INVENTION
[0039] As described herein, in some embodiments, the present invention provides methods of treating, stabilizing or lessening the severity or progression of one or more diseases or disorders associated with one or both of ERK1 and ERK2 comprising administering to a patient in need thereof an inhibitor of one or both of ERK1 and ERK2 in combination with a TOR inhibitor.
[0040] In some embodiments, an inhibitor of one or both of ERK1 and ERK2 is Compound 1, or a pharmaceutically acceptable salt thereof, as described herein.
[0041] In some embodiments, a TOR inhibitor is a compound of formula I, as described herein, infra. In certain embodiments, a TOR inhibitor is any TOR inhibitor known to one of ordinary skill in the art. Such TOR inhibitors are described herein, infra. Definitions
[0042] An“alkyl” group is a saturated, partially saturated, or unsaturated straight chain or branched non-cyclic hydrocarbon having from 1 to 10 carbon atoms, typically from 1 to 8 carbons or, in some embodiments, from 1 to 6, 1 to 4, or 2 to 6 or carbon atoms. Representative alkyl groups include -methyl, -ethyl, -n-propyl, -n-butyl, -n-pentyl and -n-hexyl; while saturated branched alkyls include -isopropyl, -sec-butyl, -isobutyl, -tert-butyl, -isopentyl, 2-methylpentyl, 3-methylpentyl, 4-methylpentyl, 2,3-dimethylbutyl and the like. Examples of unsaturated alkyl groups include, but are not limited to, vinyl, allyl, -CH=CH(CH3), -CH=C(CH3)2, -C(CH3)=CH2, -C(CH3)=CH(CH3), -C(CH2CH3)=CH2, -C≡CH, -C≡C(CH3), -C≡C(CH2CH3), -CH2C≡CH, - CH2C≡C(CH3) and -CH2C≡C(CH2CH3), among others. An alkyl group can be substituted or unsubstituted. In certain embodiments, when the alkyl groups described herein are said to be “substituted,” they may be substituted with any substituent or substituents as those found in the exemplary compounds and embodiments disclosed herein, as well as halogen (chloro, iodo, bromo, or fluoro); hydroxyl; alkoxy; alkoxyalkyl; amino; alkylamino; carboxy; nitro; cyano; thiol; thioether; imine; imide; amidine; guanidine; enamine; aminocarbonyl; acylamino; phosphonate; phosphine; thiocarbonyl; sulfinyl; sulfone; sulfonamide; ketone; aldehyde; ester; urea; urethane; oxime; hydroxyl amine; alkoxyamine; aralkoxyamine; N-oxide; hydrazine; hydrazide; hydrazone; azide; isocyanate; isothiocyanate; cyanate; thiocyanate; B(OH)2, or O(alkyl)aminocarbonyl.
[0043] An“alkenyl” group is a straight chain or branched non-cyclic hydrocarbon having from 2 to 10 carbon atoms, typically from 2 to 8 carbon atoms, and including at least one carbon- carbon double bond. Representative straight chain and branched (C2-C8)alkenyls include -vinyl, -allyl, -1-butenyl, -2-butenyl, -isobutylenyl, -1-pentenyl, -2-pentenyl, -3-methyl-1-butenyl, -2- methyl-2-butenyl, -2,3-dimethyl-2-butenyl, -1-hexenyl, -2-hexenyl, -3-hexenyl, -1-heptenyl, -2- heptenyl, -3-heptenyl, -1-octenyl, -2-octenyl, -3-octenyl and the like. The double bond of an alkenyl group can be unconjugated or conjugated to another unsaturated group. An alkenyl group can be unsubstituted or substituted.
[0044] A“cycloalkyl” group is a saturated, or partially saturated cyclic alkyl group of from 3 to 10 carbon atoms having a single cyclic ring or multiple condensed or bridged rings which can be optionally substituted with from 1 to 3 alkyl groups. In some embodiments, the cycloalkyl group has 3 to 8 ring members, whereas in other embodiments the number of ring carbon atoms ranges from 3 to 5, 3 to 6, or 3 to 7. Such cycloalkyl groups include, by way of example, single ring structures such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, 1-methylcyclopropyl, 2-methylcyclopentyl, 2-methylcyclooctyl, and the like, or multiple or bridged ring structures such as adamantyl and the like. Examples of unsaturated cycloalkyl groups include cyclohexenyl, cyclopentenyl, cyclohexadienyl, butadienyl, pentadienyl, hexadienyl, among others. A cycloalkyl group can be substituted or unsubstituted. Such substituted cycloalkyl groups include, by way of example, cyclohexanone and the like.
[0045] An“aryl” group is an aromatic carbocyclic group of from 6 to 14 carbon atoms having a single ring (e.g., phenyl) or multiple condensed rings (e.g., naphthyl or anthryl). In some embodiments, aryl groups contain 6-14 carbons, and in others from 6 to 12 or even 6 to 10 carbon atoms in the ring portions of the groups. Particular aryls include phenyl, biphenyl, naphthyl and the like. An aryl group can be substituted or unsubstituted. The phrase“aryl groups” also includes groups containing fused rings, such as fused aromatic-aliphatic ring systems (e.g., indanyl, tetrahydronaphthyl, and the like). [0046] A“heteroaryl” group is an aryl ring system having one to four heteroatoms as ring atoms in a heteroaromatic ring system, wherein the remainder of the atoms are carbon atoms. In some embodiments, heteroaryl groups contain 5 to 6 ring atoms, and in others from 6 to 9 or even 6 to 10 atoms in the ring portions of the groups. Suitable heteroatoms include oxygen, sulfur and nitrogen. In certain embodiments, the heteroaryl ring system is monocyclic or bicyclic. Non-limiting examples include but are not limited to, groups such as pyrrolyl, pyrazolyl, imidazolyl, triazolyl, tetrazolyl, oxazolyl, isoxazolyl, thiazolyl, pyrrolyl, pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl, thiophenyl, benzothiophenyl, furanyl, benzofuranyl (for example, isobenzofuran-1,3-diimine), indolyl, azaindolyl (for example, pyrrolopyridyl or 1H- pyrrolo[2,3-b]pyridyl), indazolyl, benzimidazolyl (for example, 1H-benzo[d]imidazolyl), imidazopyridyl (for example, azabenzimidazolyl, 3H-imidazo[4,5-b]pyridyl or 1H-imidazo[4,5- b]pyridyl), pyrazolopyridyl, triazolopyridyl, benzotriazolyl, benzoxazolyl, benzothiazolyl, benzothiadiazolyl, isoxazolopyridyl, thianaphthalenyl, purinyl, xanthinyl, adeninyl, guaninyl, quinolinyl, isoquinolinyl, tetrahydroquinolinyl, quinoxalinyl, and quinazolinyl groups.
[0047] A“heterocyclyl” is an aromatic (also referred to as heteroaryl) or non-aromatic cycloalkyl in which one to four of the ring carbon atoms are independently replaced with a heteroatom from the group consisting of O, S and N. In some embodiments, heterocyclyl groups include 3 to 10 ring members, whereas other such groups have 3 to 5, 3 to 6, or 3 to 8 ring members. Heterocyclyls can also be bonded to other groups at any ring atom (i.e., at any carbon atom or heteroatom of the heterocyclic ring). A heterocyclylalkyl group can be substituted or unsubstituted. Heterocyclyl groups encompass unsaturated, partially saturated and saturated ring systems, such as, for example, imidazolyl, imidazolinyl and imidazolidinyl groups. The phrase heterocyclyl includes fused ring species, including those comprising fused aromatic and non- aromatic groups, such as, for example, benzotriazolyl, 2,3-dihydrobenzo[l,4]dioxinyl, and benzo[l,3]dioxolyl. The phrase also includes bridged polycyclic ring systems containing a heteroatom such as, but not limited to, quinuclidinyl. Representative examples of a heterocyclyl group include, but are not limited to, aziridinyl, azetidinyl, pyrrolidyl, imidazolidinyl, pyrazolidinyl, thiazolidinyl, tetrahydrothiophenyl, tetrahydrofuranyl, dioxolyl, furanyl, thiophenyl, pyrrolyl, pyrrolinyl, imidazolyl, imidazolinyl, pyrazolyl, pyrazolinyl, triazolyl, tetrazolyl, oxazolyl, isoxazolyl, thiazolyl, thiazolinyl, isothiazolyl, thiadiazolyl, oxadiazolyl, piperidyl, piperazinyl, morpholinyl, thiomorpholinyl, tetrahydropyranyl (for example, tetrahydro-2H-pyranyl), tetrahydrothiopyranyl, oxathiane, dioxyl, dithianyl, pyranyl, pyridyl, pyrimidinyl, pyridazinyl, pyrazinyl, triazinyl, dihydropyridyl, dihydrodithiinyl, dihydrodithionyl, homopiperazinyl, quinuclidinyl, indolyl, indolinyl, isoindolyl, azaindolyl (pyrrolopyridyl), indazolyl, indolizinyl, benzotriazolyl, benzimidazolyl, benzofuranyl, benzothiophenyl, benzothiazolyl, benzoxadiazolyl, benzoxazinyl, benzodithiinyl, benzoxathiinyl, benzothiazinyl, benzoxazolyl, benzothiazolyl, benzothiadiazolyl, benzo[l,3]dioxolyl, pyrazolopyridyl, imidazopyridyl (azabenzimidazolyl; for example, 1H-imidazo[4,5-b]pyridyl, or 1H-imidazo[4,5- b]pyridin-2(3H)-onyl), triazolopyridyl, isoxazolopyridyl, purinyl, xanthinyl, adeninyl, guaninyl, quinolinyl, isoquinolinyl, quinolizinyl, quinoxalinyl, quinazolinyl, cinnolinyl, phthalazinyl, naphthyridinyl, pteridinyl, thianaphthalenyl, dihydrobenzothiazinyl, dihydrobenzofuranyl, dihydroindolyl, dihydrobenzodioxinyl, tetrahydroindolyl, tetrahydroindazolyl, tetrahydrobenzimidazolyl, tetrahydrobenzotriazolyl, tetrahydropyrrolopyridyl, tetrahydropyrazolopyridyl, tetrahydroimidazopyridyl, tetrahydrotriazolopyridyl, and tetrahydroquinolinyl groups. Representative substituted heterocyclyl groups may be mono- substituted or substituted more than once, such as, but not limited to, pyridyl or morpholinyl groups, which are 2-, 3-, 4-, 5-, or 6-substituted, or disubstituted with various substituents such as those listed below.
[0048] A“cycloalkylalkyl” group is a radical of the formula: -alkyl-cycloalkyl, wherein alkyl and cycloalkyl are defined above. Substituted cycloalkylalkyl groups may be substituted at the alkyl, the cycloalkyl, or both the alkyl and the cycloalkyl portions of the group. Representative cycloalkylalkyl groups include but are not limited to cyclopentylmethyl, cyclopentylethyl, cyclohexylmethyl, cyclohexylethyl, and cyclohexylpropyl. Representative substituted cycloalkylalkyl groups may be mono- substituted or substituted more than once.
[0049] An“aralkyl” group is a radical of the formula: -alkyl-aryl, wherein alkyl and aryl are defined above. Substituted aralkyl groups may be substituted at the alkyl, the aryl, or both the alkyl and the aryl portions of the group. Representative aralkyl groups include but are not limited to benzyl and phenethyl groups and fused (cycloalkylaryl)alkyl groups such as 4-ethyl-indanyl.
[0050] A“heterocyclylalkyl” group is a radical of the formula: -alkyl-heterocyclyl, wherein alkyl and heterocyclyl are defined above. Substituted heterocyclylalkyl groups may be substituted at the alkyl, the heterocyclyl, or both the alkyl and the heterocyclyl portions of the group. Representative heterocyclylalkyl groups include but are not limited to 4-ethyl- morpholinyl, 4-propylmorpholinyl, furan-2-yl methyl, furan-3-yl methyl, pyridine-3-yl methyl, (tetrahydro-2H-pyran-4-yl)methyl, (tetrahydro-2H-pyran-4-yl)ethyl, tetrahydrofuran-2-yl methyl, tetrahydrofuran-2-yl ethyl, and indol-2-yl propyl.
[0051] A“halogen” is chloro, iodo, bromo, or fluoro.
[0052] A“hydroxyalkyl” group is an alkyl group as described above substituted with one or more hydroxy groups.
[0053] An“alkoxy” group is -O-(alkyl), wherein alkyl is defined above.
[0054] An“alkoxyalkyl” group is -(alkyl)-O-(alkyl), wherein alkyl is defined above.
[0055] An“amine” group is a radical of the formula: -NH2.
[0056] A“hydroxyl amine” group is a radical of the formula: -N(R#)OH or -NHOH, wherein R# is a substituted or unsubstituted alkyl, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, heterocyclyl or heterocyclylalkyl group as defined herein.
[0057] An“alkoxyamine” group is a radical of the formula: -N(R#)O-alkyl or -NHO-alkyl, wherein R# is as defined above.
[0058] An“aralkoxyamine” group is a radical of the formula: -N(R#)O-aryl or -NHO-aryl, wherein R# is as defined above.
[0059] An“alkylamine” group is a radical of the formula: -NH-alkyl or -N(alkyl)2, wherein each alkyl is independently as defined above.
[0060] An“aminocarbonyl” group is a radical of the formula: -C(=O)N(R#)2, -C(=O)NH(R#) or -C(=O)NH2, wherein each R# is as defined above.
[0061] An “acylamino” group is a radical of the formula: -NHC(=O)(R#) or -N(alkyl)C(=O)(R#), wherein each alkyl and R# are independently as defined above.
[0062] An “O(alkyl)aminocarbonyl” group is a radical of the formula: -O(alkyl)C(=O)N(R#)2, -O(alkyl)C(=O)NH(R#) or -O(alkyl)C(=O)NH2, wherein each R# is independently as defined above.
[0063] An“N-oxide” group is a radical of the formula: -N+-O-.
[0064] A“carboxy” group is a radical of the formula: -C(=O)OH. [0065] A“ketone” group is a radical of the formula: -C(=O)(R#), wherein R# is as defined above.
[0066] An“aldehyde” group is a radical of the formula: -CH(=O).
[0067] An“ester” group is a radical of the formula: -C(=O)O(R#) or -OC(=O)(R#), wherein R# is as defined above.
[0068] A “urea” group is a radical of the formula: -N(alkyl)C(=O)N(R#)2, -N(alkyl)C(=O)NH(R#), -N(alkyl)C(=O)NH2, -NHC(=O)N(R#)2, -NHC(=O)NH(R#), or -NHC(=O)NH #
2 , wherein each alkyl and R# are independently as defined above.
[0069] An“imine” group is a radical of the formula: -N=C(R#)2 or -C(R#)=N(R#), wherein each R# is independently as defined above.
[0070] An “imide” group is a radical of the formula: -C(=O)N(R#)C(=O)(R#) or -N((C=O)(R#))2, wherein each R# is independently as defined above.
[0071] A“urethane” group is a radical of the formula: -OC(=O)N(R#)2, -OC(=O)NH(R#), - N(R#)C(=O)O(R#), or -NHC(=O)O(R#), wherein each R# is independently as defined above.
[0072] An “amidine” group is a radical of the formula: -C(=N(R#))N(R#)2, -C(=N(R#))NH(R#), -C(=N(R#))NH2, -C(=NH)N(R#)2, -C(=NH)NH(R#), -C(=NH)NH2, -N=C(R#)N(R#)2, -N=C(R#)NH(R#), -N=C(R#)NH2, -N(R#)C(R#)=N(R#), -NHC(R#)=N(R#), -N(R#)C(R#)=NH, or -NHC(R#)=NH, wherein each R# is independently as defined above.
[0073] A “guanidine” group is a radical of the formula: -N(R#)C(=N(R#))N(R#)2, -NHC(=N(R#))N(R#)2, -N(R#)C(=NH)N(R#)2, -N(R#)C(=N(R#))NH(R#), -N(R#)C(=N(R#))NH2, -NHC(=NH)N(R#)2, -NHC(=N(R#))NH(R#), -NHC(=N(R#))NH2, -NHC(=NH)NH(R#), -NHC(=NH)NH2, -N=C(N(R#)2)2, -N=C(NH(R#))2, or -N=C(NH2)2, wherein each R# is independently as defined above.
[0074] A “enamine” group is a radical of the formula: -N(R#)C(R#)=C(R#)2, -NHC(R#)=C(R#)2, -C(N(R#)2)=C(R#)2, -C(NH(R#))=C(R#)2, -C(NH2)=C(R#)2, -C(R#)=C(R#)(N(R#)2), -C(R#)=C(R#)(NH(R#)) or -C(R#)=C(R#)(NH2), wherein each R# is independently as defined above.
[0075] An“oxime” group is a radical of the formula: -C(=NO(R#))(R#), -C(=NOH)(R#), -CH(=NO(R#)), or -CH(=NOH), wherein each R# is independently as defined above. [0076] A “hydrazide” group is a radical of the formula: -C(=O)N(R#)N(R#)2, -C(=O)NHN(R#)2, -C(=O)N(R#)NH(R#), -C(=O)N(R#)NH2, -C(=O)NHNH(R#)2, or -C(=O)NHNH2, wherein each R# is independently as defined above.
[0077] A “hydrazine” group is a radical of the formula: -N(R#)N(R#)2, -NHN(R#)2, -N(R#)NH(R#), -N(R#)NH2, -NHNH(R#)2, or -NHNH2, wherein each R# is independently as defined above.
[0078] A “hydrazone” group is a radical of the formula: -C(=N-N(R#)2)(R#)2, -C(=N-NH(R#))(R#)2, -C(=N-NH2)(R#)2, -N(R#)(N=C(R#)2), or -NH(N=C(R#)2), wherein each R# is independently as defined above.
[0079] An“azide” group is a radical of the formula: -N3.
[0080] An“isocyanate” group is a radical of the formula: -N=C=O.
[0081] An“isothiocyanate” group is a radical of the formula: -N=C=S.
[0082] A“cyanate” group is a radical of the formula: -OCN.
[0083] A“thiocyanate” group is a radical of the formula: -SCN.
[0084] A“thioether” group is a radical of the formula; -S(R#), wherein R# is as defined above.
[0085] A“thiocarbonyl” group is a radical of the formula: -C(=S)(R#), wherein R# is as defined above.
[0086] A“sulfinyl” group is a radical of the formula: -S(=O)(R#), wherein R# is as defined above.
[0087] A“sulfone” group is a radical of the formula: -S(=O)2(R#), wherein R# is as defined above.
[0088] A “sulfonylamino” group is a radical of the formula: -NHSO2(R#) or -N(alkyl)SO2(R#), wherein each alkyl and R# are defined above.
[0089] A“sulfonamide” group is a radical of the formula: -S(=O)2N(R#)2, or -S(=O)2NH(R#), or -S(=O)2NH2, wherein each R# is independently as defined above.
[0090] A“phosphonate” group is a radical of the formula: -P(=O)(O(R#))2, -P(=O)(OH)2, -OP(=O)(O(R#))(R#), or -OP(=O)(OH)(R#), wherein each R# is independently as defined above.
[0091] A“phosphine” group is a radical of the formula: -P(R#)2, wherein each R# is independently as defined above. [0092] When the groups described herein, with the exception of alkyl group are said to be “substituted,” they may be substituted with any appropriate substituent or substituents. Illustrative examples of substituents are those found in the exemplary compounds and embodiments disclosed herein, as well as halogen (chloro, iodo, bromo, or fluoro); alkyl; hydroxyl; alkoxy; alkoxyalkyl; amino; alkylamino; carboxy; nitro; cyano; thiol; thioether; imine; imide; amidine; guanidine; enamine; aminocarbonyl; acylamino; phosphonate; phosphine; thiocarbonyl; sulfinyl; sulfone; sulfonamide; ketone; aldehyde; ester; urea; urethane; oxime; hydroxyl amine; alkoxyamine; aralkoxyamine; N-oxide; hydrazine; hydrazide; hydrazone; azide; isocyanate; isothiocyanate; cyanate; thiocyanate; oxygen (═O); B(OH)2, O(alkyl)aminocarbonyl; cycloalkyl, which may be monocyclic or fused or non-fused polycyclic (e.g., cyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl), or a heterocyclyl, which may be monocyclic or fused or non-fused polycyclic (e.g., pyrrolidyl, piperidyl, piperazinyl, morpholinyl, or thiazinyl); monocyclic or fused or non-fused polycyclic aryl or heteroaryl (e.g., phenyl, naphthyl, pyrrolyl, indolyl, furanyl, thiophenyl, imidazolyl, oxazolyl, isoxazolyl, thiazolyl, triazolyl, tetrazolyl, pyrazolyl, pyridinyl, quinolinyl, isoquinolinyl, acridinyl, pyrazinyl, pyridazinyl, pyrimidinyl, benzimidazolyl, benzothiophenyl, or benzofuranyl) aryloxy; aralkyloxy; heterocyclyloxy; and heterocyclyl alkoxy.
[0093] As used herein, the term“pharmaceutically acceptable salt(s)” refers to a salt prepared from a pharmaceutically acceptable non-toxic acid or base including an inorganic acid and base and an organic acid and base. Suitable pharmaceutically acceptable base addition salts include, but are not limited to metallic salts made from aluminum, calcium, lithium, magnesium, potassium, sodium and zinc or organic salts made from lysine, N,N’-dibenzylethylenediamine, chloroprocaine, choline, diethanolamine, ethylenediamine, meglumine (N-methylglucamine) and procaine. Suitable non-toxic acids include, but are not limited to, inorganic and organic acids such as acetic, alginic, anthranilic, benzenesulfonic or besylate, benzoic, camphorsulfonic, citric, ethenesulfonic, formic, fumaric, furoic, galacturonic, gluconic, glucuronic, glutamic, glycolic, hydrobromic, hydrochloric, isethionic, lactic, maleic, malic, mandelic, methanesulfonic, mucic, nitric, pamoic, pantothenic, phenylacetic, phosphoric, propionic, salicylic, stearic, succinic, sulfanilic, sulfuric, tartaric acid, and p-toluenesulfonic acid. Specific non-toxic acids include hydrochloric, hydrobromic, phosphoric, sulfuric, and methanesulfonic acids. Examples of specific salts thus include hydrochloride and mesylate salts. Others are well-known in the art, see for example, Remington’s Pharmaceutical Sciences, 18th eds., Mack Publishing, Easton PA (1990) or Remington: The Science and Practice of Pharmacy, 19th eds., Mack Publishing, Easton PA (1995).
[0094] As used herein and unless otherwise indicated, the term“clathrate” means a TOR inhibitor, 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 or a crystal lattice wherein a TOR inhibitor is a guest molecule.
[0095] As used herein and unless otherwise indicated, the term“solvate” means a TOR inhibitor, or a salt thereof, that further includes a stoichiometric or non-stoichiometric amount of a solvent bound by non-covalent intermolecular forces. In one embodiment, the solvate is a hydrate.
[0096] As used herein and unless otherwise indicated, the term“hydrate” means a TOR inhibitor, or a salt thereof, that further includes a stoichiometric or non-stoichiometric amount of water bound by non-covalent intermolecular forces.
[0097] As used herein and unless otherwise indicated, the term“prodrug” means a TOR inhibitor derivative that can hydrolyze, oxidize, or otherwise react under biological conditions (in vitro or in vivo) to provide an active compound, particularly a TOR inhibitor. Examples of prodrugs include, but are not limited to, derivatives and metabolites of a TOR inhibitor that include biohydrolyzable moieties such as biohydrolyzable amides, biohydrolyzable esters, biohydrolyzable carbamates, biohydrolyzable carbonates, biohydrolyzable ureides, and biohydrolyzable phosphate analogues. In certain embodiments, prodrugs of compounds with carboxyl functional groups are the lower alkyl esters of the carboxylic acid. The carboxylate esters are conveniently formed by esterifying any of the carboxylic acid moieties present on the molecule. Prodrugs can typically be prepared using well-known methods, such as those described by Burger’s Medicinal Chemistry and Drug Discovery 6th ed. (Donald J. Abraham ed., 2001, Wiley) and Design and Application of Prodrugs (H. Bundgaard ed., 1985, Harwood Academic Publishers Gmfh).
[0098] As used herein and unless otherwise indicated, the term “stereoisomer” or “stereomerically pure” means one stereoisomer of a TOR inhibitor that is substantially free of other stereoisomers of that compound. For example, a stereomerically pure compound having one chiral center will be substantially free of the opposite enantiomer of the compound. A stereomerically pure compound having two chiral centers will be substantially free of other diastereomers of the compound. A typical stereomerically pure compound comprises greater than about 80% by weight of one stereoisomer of the compound and less than about 20% by weight of other stereoisomers of the compound, greater than about 90% by weight of one stereoisomer of the compound and less than about 10% by weight of the other stereoisomers of the compound, greater than about 95% by weight of one stereoisomer of the compound and less than about 5% by weight of the other stereoisomers of the compound, or greater than about 97% by weight of one stereoisomer of the compound and less than about 3% by weight of the other stereoisomers of the compound. The TOR inhibitors can have chiral centers and can occur as racemates, individual enantiomers or diastereomers, and mixtures thereof. All such isomeric forms are included within the embodiments disclosed herein, including mixtures thereof. The use of stereomerically pure forms of such TOR inhibitors, as well as the use of mixtures of those forms are encompassed by the embodiments disclosed herein. For example, mixtures comprising equal or unequal amounts of the enantiomers of a particular TOR inhibitor may be used in methods and compositions disclosed herein. These isomers may be asymmetrically synthesized or resolved using standard techniques such as chiral columns or chiral resolving agents. See, e.g., Jacques, J., et al., Enantiomers, Racemates and Resolutions (Wiley-Interscience, New York, 1981); Wilen, S. H., et al., Tetrahedron 33:2725 (1977); Eliel, E. L., Stereochemistry of Carbon Compounds (McGraw-Hill, NY, 1962); and Wilen, S. H., Tables of Resolving Agents and Optical Resolutions p. 268 (E.L. Eliel, Ed., Univ. of Notre Dame Press, Notre Dame, IN, 1972).
[0099] It should also be noted the TOR inhibitors can include E and Z isomers, or a mixture thereof, and cis and trans isomers or a mixture thereof. In certain embodiments, the TOR inhibitors are isolated as either the cis or trans isomer. In other embodiments, the TOR inhibitors are a mixture of the cis and trans isomers.
[00100] “Tautomers” refers to isomeric forms of a compound that are in equilibrium with each other. The concentrations of the isomeric forms will depend on the environment the compound is found in and may be different depending upon, for example, whether the compound is a solid or is in an organic or aqueous solution. For example, in aqueous solution, pyrazoles may exhibit the following isomeric forms, which are referred to as tautomers of each other:
Figure imgf000018_0001
[00101] As readily understood by one skilled in the art, a wide variety of functional groups and other structures may exhibit tautomerism and all tautomers of the TOR inhibitors are within the scope of the present invention.
[00102] It should also be noted the TOR inhibitors can contain unnatural proportions of atomic isotopes at one or more of the atoms. For example, the compounds may be radiolabeled with radioactive isotopes, such as for example tritium (3H), iodine-125 (125I), sulfur-35 (35S), or carbon-14 (14C), or may be isotopically enriched, such as with deuterium (2H), carbon-13 (13C), or nitrogen-15 (15N). As used herein, an“isotopologue” is an isotopically enriched compound. The term“isotopically enriched” refers to an atom having an isotopic composition other than the natural isotopic composition of that atom.“Isotopically enriched” may also refer to a compound containing at least one atom having an isotopic composition other than the natural isotopic composition of that atom. The term“isotopic composition” refers to the amount of each isotope present for a given atom. Radiolabeled and isotopically enriched compounds are useful as therapeutic agents, e.g., cancer and inflammation therapeutic agents, research reagents, e.g., binding assay reagents, and diagnostic agents, e.g., in vivo imaging agents. All isotopic variations of the TOR inhibitors as described herein, whether radioactive or not, are intended to be encompassed within the scope of the embodiments provided herein. In some embodiments, there are provided isotopologues of the TOR inhibitors, for example, the isotopologues are deuterium, carbon-13, or nitrogen-15 enriched TOR inhibitors.
It should be noted that if there is a discrepancy between a depicted structure and a name for that structure, the depicted structure is to be accorded more weight.
[00103] As used herein, a“disease or disorder associated with one or both of ERK1 and ERK2” means any disease or other deleterious condition in which one or both of ERK1 and ERK2, or a mutant thereof, is known or suspected to play a role. As described further herein, one of ordinary skill in the art will appreciate that ERK1 and ERK2 are downstream targets within the MAPK pathway. Thus, without wishing to be bound by any particular theory, a disease or disorder associated with one or both of ERK1 and ERK2 includes those in which activation of the MAPK pathway at any level (Ras-Raf-Mek-ERK) is known or suspected to play a role, including one or both of ERK1 and ERK2 as well as other nodes in the MAPK pathway upstream from ERK (such as Ras, Raf and Mek).Accordingly, another embodiment of the present invention relates to preventing, treating, stabilizing or lessening the severity or progression of one or more diseases in which one or both of ERK1 and ERK2, or a mutant thereof, is known or suspected to play a role. In some embodiments, the present invention relates to a method of treating or lessening the severity of a proliferative disorder, wherein said method comprises administering to a patient in need thereof Compound 1 in combination with a TOR inhibitor.
[00104] As used herein, the term“irreversible” or“irreversible inhibitor” refers to an inhibitor (i.e. a compound) that is able to be covalently bonded to a target protein kinase in a substantially non-reversible manner. That is, whereas a reversible inhibitor is able to bind to (but is generally unable to form a covalent bond to) the target protein kinase, and therefore can become dissociated from the target protein kinase, an irreversible inhibitor will remain substantially bound to the target protein kinase once covalent bond formation has occurred. Irreversible inhibitors usually display time dependency, whereby the degree of inhibition increases with the time with which the inhibitor is in contact with the enzyme. Methods for identifying if a compound is acting as an irreversible inhibitor are known to one of ordinary skill in the art. Such methods include, but are not limited to, enzyme kinetic analysis of the inhibition profile of the compound with the protein kinase target, the use of mass spectrometry of the protein drug target modified in the presence of the inhibitor compound, discontinuous exposure, also known as“washout,” experiments, and the use of labeling, such as radiolabelled inhibitor, to show covalent modification of the enzyme, as well as other methods known to one of skill in the art.
[00105] The term“subject”, as used herein, means a mammal and includes human and animal subjects, such as domestic animals (e.g., horses, dogs, cats, etc.).
[00106] As used herein, a“therapeutically effective amount” means an amount of a substance (e.g., a therapeutic agent, composition, and/or formulation) that elicits a desired biological response. In some embodiments, a therapeutically effective amount of a substance is an amount that is sufficient, when administered as part of a dosing regimen to a subject suffering from or susceptible to a disease, condition, or disorder, to treat, diagnose, prevent, and/or delay the onset of the disease, condition, or disorder. As will be appreciated by those of ordinary skill in this art, the effective amount of a substance may vary depending on such factors as the desired biological endpoint, the substance to be delivered, the target cell or tissue, etc. For example, the effective amount of compound in a formulation to treat a disease, condition, or disorder is the amount that alleviates, ameliorates, relieves, inhibits, prevents, delays onset of, reduces severity of and/or reduces incidence of one or more symptoms or features of the disease, condition, or disorder. In some embodiments, a“therapeutically effective amount” is at least a minimal amount of a compound, or composition containing a compound, which is sufficient for treating one or more symptoms of a disease or disorder associated with one or both of ERK1 and ERK2.
[00107] The terms“treat” or“treating,” as used herein, refers to partially or completely alleviating, inhibiting, delaying onset of, preventing, ameliorating and/or relieving a disease or disorder, or one or more symptoms of the disease or disorder. As used herein, the terms “treatment,” “treat,” and“treating” refer to partially or completely alleviating, inhibiting, delaying onset of, preventing, ameliorating and/or relieving a disease or disorder, or one or more symptoms of the disease or disorder, as described herein. In some embodiments, treatment may be administered after one or more symptoms have developed. In some embodiments, the term “treating” includes preventing or halting the progression of a disease or disorder. In other embodiments, treatment may be administered in the absence of symptoms. For example, treatment may be administered to a susceptible individual prior to the onset of symptoms (e.g., in light of a history of symptoms and/or in light of genetic or other susceptibility factors). Treatment may also be continued after symptoms have resolved, for example to prevent or delay their recurrence. Thus, in some embodiments, the term“treating” includes preventing relapse or recurrence of a disease or disorder.
[00108] The expression“unit dosage form” as used herein refers to a physically discrete unit of therapeutic formulation appropriate for the subject to be treated. It will be understood, however, that the total daily usage of the compositions of the present invention will be decided by the attending physician within the scope of sound medical judgment. The specific effective dose level for any particular subject or organism will depend upon a variety of factors including the disorder being treated and the severity of the disorder; activity of specific active agent employed; specific composition employed; age, body weight, general health, sex and diet of the subject; time of administration, and rate of excretion of the specific active agent employed; duration of the treatment; drugs and/or additional therapies used in combination or coincidental with specific compound(s) employed, and like factors well known in the medical arts. Compound 1 and Pharmaceutically Acceptable Salts Thereof
[00109] As described generally above, in some embodiments, the present invention provides methods of treating, stabilizing or lessening the severity or progression of one or more diseases or disorders associated with one or both of ERK1 and ERK2 comprising administering to a patient in need thereof an inhibitor of one or both of ERK 1 and ERK2 in combination with a TOR inhibitor. In some aspects, the inhibitor of one or both of ERK1 and ERK2 is Compound 1 (N-(2-((2-((2-methoxy-5-methylpyridin-4-yl)amino)-5-(trifluoromethyl)pyrimidin-4-yl)amino)-5- methylphenyl)acrylamide):
Figure imgf000021_0001
or a pharmaceutically acceptable salt thereof. For instance, in some embodiments, Compound 1 is in the form of a phosphate salt.
[00110] Compound 1, N-(2-((2-((2-methoxy-5-methylpyridin-4-yl)amino)-5- (trifluoromethyl)pyrimidin-4-yl)amino)-5-methylphenyl)acrylamide, is designated as compound number I-90 in PCT patent application serial number PCT/US14/15256, filed February 7, 2014 and published as WO2014/124230 on August 14, 2014 (referred to herein as“the ‘230 publication,”) the entirety of which is hereby incorporated by reference. The synthesis of Compound 1 is described in detail at Example 94 of the‘230 publication. Compound 1 is active in a variety of assays and therapeutic models demonstrating covalent, irreversible inhibition of one or both of ERK1 and ERK2 kinases (see, e.g., Table A of the‘230 publication). [00111] In some embodiments, the present invention provides a method of treating, stabilizing or lessening the severity or progression of one or more diseases or disorders associated with one or both of ERK1 and ERK2, comprising administering to a patient in need thereof a composition comprising Compound 1:
Figure imgf000022_0001
or a pharmaceutically acceptable salt thereof, in combination with a TOR inhibitor.
[00112] It is understood that although the methods described herein may refer to formulations, doses and dosing regimens/schedules of Compound 1, such formulations, doses and/or dosing regimens/schedules are equally applicable to any pharmaceutically acceptable salt of Compound 1. Accordingly, in some embodiments, a dose or dosing regimen for a pharmaceutically acceptable salt of Compound 1 is selected from any of the doses or dosing regimens for Compound 1 as described herein.
[00113] As described in the‘230 publication, Compound 1 is a potent inhibitor of the kinase activities of ERK1 and ERK2. In some embodiments, Compound 1 inhibits one or both of ERK1 and ERK2 with an IC50 of about 10 to about 20 nM. Compound 1 irreversibly inhibits ERK1 and ERK2 through formation of a covalent adduct with critical cysteine residues (amino acid 183 in ERK1 and 166 in ERK2) in the vicinity of the ATP binding pocket. In an analysis of 258 kinases, Compound 1 was shown to exhibit good overall kinase selectivity profile.
[00114] Compound 1 has demonstrated potent in vitro anti-proliferative activity against a large number of cancer cell lines of various tissue origins. Bioinformatic analyses indicate that tumors with activating mutations of BRAF are particularly sensitive to Compound 1. Notably, of 27 BRAF-mutant cancer cell lines tested, 25 (93%) demonstrated sensitivity to Compound 1 inhibition (GI50 <1µM). In the same cancer cell panel screening, 28 of 37 (76%) KRAS-mutant cancer cell lines were sensitive to Compound 1. Compound 1 also exhibits inhibitory activity against, for instance, A375 melanoma cells that have acquired in vitro resistance to BRAF and MEK inhibition. This is of particular importance as resistance to BRAF inhibition has been commonly observed in human patients and described in relevant in vitro and in vivo models. Such patients whose tumors demonstrated resistance to BRAF inhibitors are often cross-resistant to MEK inhibitors. Without wishing to being bound by any particular theory, it is believed that inhibitors of one or both of ERK1 and ERK2, or a mutant thereof, such as Compound 1, or pharmaceutically acceptable salts thereof, provide effective salvage therapy. TOR Inhibitors
[00115] As described generally above, provided methods comprise combination therapies utilizing an inhibitor of one or both of ERK1 and ERK2 and a TOR inhibitor. As used herein, the term“TOR inhibitor” includes mTOR kinase inhibitors and allosteric mTOR inhibitors. Examples of both mTOR kinase inhibitors and allosteric mTOR inhibitors are known to one of ordinary skill in the art and include those described herein, infra.
[00116] The protein named mTOR (mammalian target of rapamycin), also known as FRAP, RAFTI, or RAPT1, is a Ser/Thr protein kinase related to the lipid kinases of the phosphoinositide 3-kinase (PI3K) family. It functions as a sensor of mitogen, energy, and nutrient levels, and is one of the central controllers of cell growth. mTOR has been shown to be a critical protein in the PI3K/Akt/mTOR pathway that regulates cell growth and proliferation (Georgakis and Younes, Expert Rev. Anticancer Ther. 6(1):131-140 (2006)), and various mTOR inhibitors have been, or are being, evaluated in human clinical trials for the treatment of cancer.
[00117] In some embodiments, the present invention provides a method of treating, stabilizing or lessening the severity or progression of one or more diseases or disorders associated with one or both of ERK1 and ERK2, or a mutant thereof, comprising administering to a patient in need thereof Compound 1, or a pharmaceutically acceptable salt thereof, in combination with a TOR inhibitor.
[00118] In some embodiments, the present invention provides a method of treating, stabilizing or lessening the severity or progression of one or more diseases or disorders associated with one or both of ERK1 and ERK2, or a mutant thereof, comprising administering to a patient in need thereof Compound 1, or a pharmaceutically acceptable salt thereof, in combination with an mTOR kinase inhibitor. [00119] In some embodiments, the present invention provides a method of treating, stabilizing or lessening the severity or progression of one or more diseases or disorders associated with one or both of ERK1 and ERK2, or a mutant thereof, comprising administering to a patient in need thereof Compound 1, or a pharmaceutically acceptable salt thereof, in combination with an allosteric mTOR inhibitor.
[00120] In some embodiments, Compound 1, or a pharmaceutically acceptable salt thereof, is administered in combination with a TOR inhibitor of formula (I), depicted below:
Figure imgf000024_0001
and pharmaceutically acceptable salts, clathrates, solvates, stereoisomers, tautomers, metabolites, isotopologues and prodrugs thereof, wherein:
R1 is substituted or unsubstituted C1-8 alkyl, substituted or unsubstituted aryl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocyclyl, or substituted or unsubstituted heterocyclylalkyl;
R2 is H, substituted or unsubstituted C1-8 alkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocyclyl, substituted or unsubstituted heterocyclylalkyl, substituted or unsubstituted aralkyl, or substituted or unsubstituted cycloalkylalkyl; and
R3 is H, or a substituted or unsubstituted C1-8 alkyl.
[00121] In some embodiments of compounds of formula (I), R1 is substituted or unsubstituted aryl or substituted or unsubstituted heteroaryl. For example, R1 is phenyl, pyridyl, pyrimidyl, benzimidazolyl, 1H-pyrrolo[2,3-b]pyridyl, indazolyl, indolyl, 1H-imidazo[4,5-b]pyridyl, 1H- imidazo[4,5-b]pyridin-2(3H)-onyl, 3H-imidazo[4,5-b]pyridyl, or pyrazolyl, each optionally substituted. In some embodiments, R1 is phenyl substituted with one or more substituents independently selected from the group consisting of substituted or unsubstituted C1-8 alkyl (for example, methyl), substituted or unsubstituted heterocyclyl (for example, a substituted or unsubstituted triazolyl or pyrazolyl), aminocarbonyl, halogen (for example, fluorine), cyano, hydroxyalkyl and hydroxy. In other embodiments, R1 is pyridyl substituted with one or more substituents independently selected from the group consisting of substituted or unsubstituted C1-8 alkyl (for example, methyl), substituted or unsubstituted heterocyclyl (for example, a substituted or unsubstituted triazolyl), halogen, aminocarbonyl , cyano, hydroxyalkyl (for example, hydroxypropyl), -OR, and -NR2, wherein each R is independently H, or a substituted or unsubstituted C1-4 alkyl. In some embodiments, R1 is 1H-pyrrolo[2,3-b]pyridyl or benzimidazolyl, optionally substituted with one or more substituents independently selected from the group consisting of substituted or unsubstituted C1-8 alkyl, and -NR2, wherein R is independently H, or a substituted or unsubstituted C1-4 alkyl.
[00122] In some embodiments, R1 is
Figure imgf000025_0001
wherein R is at each occurrence independently H, or a substituted or unsubstituted C1-4 alkyl (for example, methyl); R’ is at each occurrence independently a substituted or unsubstituted C1-4 alkyl (for example, methyl), halogen (for example, fluoro), cyano, -OR, or - NR2; m is 0-3; and n is 0-3. It will be understood by those skilled in the art that any of the subsituuents R’ may be attached to any suitable atom of any of the rings in the fused ring systems.
[00123] In some embodiments of compounds of formula (I), R1 is
Figure imgf000026_0001
wherein R is at each occurrence independently H, or a substituted or unsubstituted C1-4 alkyl; R’ is at each occurrence independently a substituted or unsubstituted C1-4 alkyl, halogen, cyano, -OR or -NR2; m is 0-3; and n is 0-3.
[00124] In some embodiments of compounds of formula (I), R2 is H, substituted or unsubstituted C1-8 alkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocyclyl, substituted or unsubstituted C1-4 alkyl-heterocyclyl, substituted or unsubstituted C1- 4 alkyl-aryl, or substituted or unsubstituted C1-4 alkyl-cycloalkyl. For example, R2 is H, methyl, ethyl, n-propyl, isopropyl, n-butyl, sec-butyl, isobutyl, tert-butyl, n-pentyl, isopentyl, cyclopentyl, cyclohexyl, tetrahydrofuranyl, tetrahydropyranyl, (C1-4 alkyl)-phenyl, (C1-4 alkyl)- cyclopropyl, (C1-4 alkyl)-cyclobutyl, (C1-4 alkyl)-cyclopentyl, (C1-4 alkyl)-cyclohexyl, (C1-4 alkyl)-pyrrolidyl, (C1-4 alkyl)-piperidyl, (C1-4 alkyl)-piperazinyl, (C1-4 alkyl)-morpholinyl, (C1-4 alkyl)-tetrahydrofuranyl, or (C1-4 alkyl)-tetrahydropyranyl, each optionally substituted.
[00125] In other embodiments, R2 is H, C1-4 alkyl, (C1-4 alkyl)(OR),
Figure imgf000027_0001
wherein R is at each occurrence independently H, or a substituted or unsubstituted C1-4 alkyl (for example, methyl); R’ is at each occurrence independently H, -OR, cyano, or a substituted or unsubstituted C1-4 alkyl (for example, methyl); and p is 0-3.
[00126] In other embodiments of compounds of formula (I), R2 is H, C1-4 alkyl, (C1-4alkyl)(OR),
Figure imgf000027_0002
wherein R is at each occurrence independently H, or a substituted or unsubstituted C1-2 alkyl; R’ is at each occurrence independently H, -OR, cyano, or a substituted or unsubstituted C1-2 alkyl; and p is 0-1.
[00127] In some embodiments of compounds of formula (I), R3 is H.
[00128] In some such embodiments described herein, R1 is substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl. For example, R1 is phenyl, pyridyl, pyrimidyl, benzimidazolyl, 1H-pyrrolo[2,3-b]pyridyl, indazolyl, indolyl, 1H-imidazo[4,5-b]pyridine, pyridyl, 1H-imidazo[4,5-b]pyridin-2(3H)-onyl, 3H-imidazo[4,5-b]pyridyl, or pyrazolyl, each optionally substituted. In some embodiments, R1 is phenyl substituted with one or more substituents independently selected from the group consisting of substituted or unsubstituted C1-8 alkyl, substituted or unsubstituted heterocyclyl, aminocarbonyl, halogen, cyano, hydroxyalkyl and hydroxy. In others, R1 is pyridyl substituted with one or more substituents independently selected from the group consisting of C1-8 alkyl, substituted or unsubstituted heterocyclyl, halogen, aminocarbonyl, cyano, hydroxyalkyl, -OR, and -NR2, wherein each R is independently H, or a substituted or unsubstituted C1-4 alkyl. In still others, R1 is 1H- pyrrolo[2,3-b]pyridyl or benzimidazolyl, optionally substituted with one or more substituents independently selected from the group consisting of substituted or unsubstituted C1-8 alkyl, and -NR2, wherein R is independently H, or a substituted or unsubstituted C1-4 alkyl.
[00129] In certain embodiments, the compounds of formula (I) have an R1 group set forth herein and an R2 group set forth herein.
[00130] In some embodiments of compounds of formula (I), the compound at a concentration of 10 μM inhibits mTOR, DNA-PK, PI3K, or a combination thereof by at least about 50%. Compounds of formula (I) may be shown to be inhibitors of the kinases above in any suitable assay system.
[00131] Representative TOR inhibitors of formula (I) include compounds from Table A.
Table A.
7-(5-fluoro-2-methyl-4-(1H-1,2,4-triazol-3-yl)phenyl)-1-((trans-4-methoxycyclohexyl)methyl)- 3,4-dihydropyrazino[2,3-b]pyrazin-2(1H)-one;
7-(6-(1H-1,2,4-triazol-3-yl)pyridin-3-yl)-1-(cis-4-methoxycyclohexyl)-3,4-dihydropyrazino[2,3- b]pyrazin-2(1H)-one;
7-(1H-pyrrolo[2,3-b]pyridin-3-yl)-1-(2-(tetrahydro-2H-pyran-4-yl)ethyl)-3,4- dihydropyrazino[2,3-b]pyrazin-2(1H)-one;
7-(5-fluoro-2-methyl-4-(1H-1,2,4-triazol-3-yl)phenyl)-1-((cis-4-methoxycyclohexyl)methyl)- 3,4-dihydropyrazino[2,3-b]pyrazin-2(1H)-one;
1-ethyl-7-(1H-pyrrolo[3,2-b]pyridin-5-yl)-3,4-dihydropyrazino[2,3-b]pyrazin-2(1H)-one;
7-(6-(1H-1,2,4-triazol-3-yl)pyridin-3-yl)-1-((cis-4-methoxycyclohexyl)methyl)-3,4- dihydropyrazino[2,3-b]pyrazin-2(1H)-one;
7-(1H-benzo[d]imidazol-4-yl)-1-(2-(tetrahydro-2H-pyran-4-yl)ethyl)-3,4-dihydropyrazino[2,3- b]pyrazin-2(1H)-one; 7-(1H-pyrrolo[2,3-b]pyridin-4-yl)-1-(2-(tetrahydro-2H-pyran-4-yl)ethyl)-3,4- dihydropyrazino[2,3-b]pyrazin-2(1H)-one;
7-(6-(1H-1,2,4-triazol-3-yl)pyridin-3-yl)-1-((trans-4-methoxycyclohexyl)methyl)-3,4- dihydropyrazino[2,3-b]pyrazin-2(1H)-one;
7-(6-(1H-1,2,4-triazol-3-yl)pyridin-3-yl)-1-((trans-4-hydroxycyclohexyl)methyl)-3,4- dihydropyrazino[2,3-b]pyrazin-2(1H)-one;
7-(6-(1H-1,2,4-triazol-3-yl)pyridin-3-yl)-1-(cis-4-hydroxycyclohexyl)-3,4-dihydropyrazino[2,3- b]pyrazin-2(1H)-one;
7-(5-fluoro-2-methyl-4-(1H-1,2,4-triazol-3-yl)phenyl)-1-(cis-4-hydroxycyclohexyl)-3,4- dihydropyrazino[2,3-b]pyrazin-2(1H)-one;
7-(6-(1H-1,2,4-triazol-3-yl)pyridin-3-yl)-1-(tetrahydro-2H-pyran-4-yl)-3,4-dihydropyrazino[2,3- b]pyrazin-2(1H)-one;
7-(6-(1H-1,2,4-triazol-3-yl)pyridin-3-yl)-1-(2-methoxyethyl)-3,4-dihydropyrazino[2,3-b]pyrazin- 2(1H)-one;
7-(6-(1H-1,2,4-triazol-3-yl)pyridin-3-yl)-1-ethyl-3,4-dihydropyrazino[2,3-b]pyrazin-2(1H)-one; 7-(5-fluoro-2-methyl-4-(1H-1,2,4-triazol-3-yl)phenyl)-1-((cis-4-hydroxycyclohexyl)methyl)-3,4- dihydropyrazino[2,3-b]pyrazin-2(1H)-one;
7-(5-fluoro-2-methyl-4-(1H-1,2,4-triazol-3-yl)phenyl)-1-(tetrahydro-2H-pyran-4-yl)-3,4- dihydropyrazino[2,3-b]pyrazin-2(1H)-one;
7-(1H-indol-4-yl)-1-(2-(tetrahydro-2H-pyran-4-yl)ethyl)-3,4-dihydropyrazino[2,3-b]pyrazin- 2(1H)-one;
7-(5-fluoro-2-methyl-4-(1H-1,2,4-triazol-3-yl)phenyl)-1-((trans-4-hydroxycyclohexyl)methyl)- 3,4-dihydropyrazino[2,3-b]pyrazin-2(1H)-one;
7-(6-(1H-1,2,4-triazol-3-yl)pyridin-3-yl)-1-((cis-4-hydroxycyclohexyl)methyl)-3,4- dihydropyrazino[2,3-b]pyrazin-2(1H)-one;
7-(6-(1H-1,2,4-triazol-3-yl)pyridin-3-yl)-1-(trans-4-hydroxycyclohexyl)-3,4- dihydropyrazino[2,3-b]pyrazin-2(1H)-one;
7-(6-(1H-1,2,4-triazol-3-yl)pyridin-3-yl)-1-(trans-4-methoxycyclohexyl)-3,4- dihydropyrazino[2,3-b]pyrazin-2(1H)-one; 7-(6-(1H-1,2,4-triazol-3-yl)pyridin-3-yl)-1-isopropyl-3,4-dihydropyrazino[2,3-b]pyrazin-2(1H)- one;
7-(5-fluoro-2-methyl-4-(1H-1,2,4-triazol-3-yl)phenyl)-1-(trans-4-methoxycyclohexyl)-3,4- dihydropyrazino[2,3-b]pyrazin-2(1H)-one;
7-(5-fluoro-2-methyl-4-(1H-1,2,4-triazol-3-yl)phenyl)-1-(trans-4-hydroxycyclohexyl)-3,4- dihydropyrazino[2,3-b]pyrazin-2(1H)-one;
7-(5-fluoro-2-methyl-4-(1H-1,2,4-triazol-3-yl)phenyl)-1-(2-methoxyethyl)-3,4- dihydropyrazino[2,3-b]pyrazin-2(1H)-one;
7-(5-fluoro-2-methyl-4-(1H-1,2,4-triazol-3-yl)phenyl)-1-isopropyl-3,4-dihydropyrazino[2,3- b]pyrazin-2(1H)-one;
1-ethyl-7-(5-fluoro-2-methyl-4-(1H-1,2,4-triazol-3-yl)phenyl)-3,4-dihydropyrazino[2,3- b]pyrazin-2(1H)-one;
7-(2-hydroxypyridin-4-yl)-1-(2-(tetrahydro-2H-pyran-4-yl)ethyl)-3,4-dihydropyrazino[2,3- b]pyrazin-2(1H)-one;
1-isopropyl-7-(4-methyl-6-(1H-1,2,4-triazol-3-yl)pyridin-3-yl)-3,4-dihydropyrazino[2,3- b]pyrazin-2(1H)-one;
5-(8-isopropyl-7-oxo-5,6,7,8-tetrahydropyrazino[2,3-b]pyrazin-2-yl)-4-methylpicolinamide; 7-(1H-indazol-4-yl)-1-(2-(tetrahydro-2H-pyran-4-yl)ethyl)-3,4-dihydropyrazino[2,3-b]pyrazin- 2(1H)-one;
7-(2-aminopyrimidin-5-yl)-1-(2-(tetrahydro-2H-pyran-4-yl)ethyl)-3,4-dihydropyrazino[2,3- b]pyrazin-2(1H)-one;
7-(2-aminopyridin-4-yl)-1-(2-(tetrahydro-2H-pyran-4-yl)ethyl)-3,4-dihydropyrazino[2,3- b]pyrazin-2(1H)-one;
7-(6-(methylamino)pyridin-3-yl)-1-(2-(tetrahydro-2H-pyran-4-yl)ethyl)-3,4- dihydropyrazino[2,3-b]pyrazin-2(1H)-one;
7-(6-hydroxypyridin-3-yl)-1-(2-(tetrahydro-2H-pyran-4-yl)ethyl)-3,4-dihydropyrazino[2,3- b]pyrazin-2(1H)-one;
7-(4-(1H-pyrazol-3-yl)phenyl)-1-(2-methoxyethyl)-3,4-dihydropyrazino[2,3-b]pyrazin-2(1H)- one; 7-(pyridin-3-yl)-1-(2-(tetrahydro-2H-pyran-4-yl)ethyl)-3,4-dihydropyrazino[2,3-b]pyrazin- 2(1H)-one;
7-(1H-indazol-4-yl)-1-(2-methoxyethyl)-3,4-dihydropyrazino[2,3-b]pyrazin-2(1H)-one;
7-(1H-indazol-6-yl)-1-(2-methoxyethyl)-3,4-dihydropyrazino[2,3-b]pyrazin-2(1H)-one;
7-(pyrimidin-5-yl)-1-(2-(tetrahydro-2H-pyran-4-yl)ethyl)-3,4-dihydropyrazino[2,3-b]pyrazin- 2(1H)-one;
7-(6-methoxypyridin-3-yl)-1-(2-(tetrahydro-2H-pyran-4-yl)ethyl)-3,4-dihydropyrazino[2,3- b]pyrazin-2(1H)-one;
1-(2-methoxyethyl)-7-(1H-pyrrolo[2,3-b]pyridin-5-yl)-3,4-dihydropyrazino[2,3-b]pyrazin- 2(1H)-one;
1-ethyl-7-(1H-pyrrolo[2,3-b]pyridin-5-yl)-3,4-dihydropyrazino[2,3-b]pyrazin-2(1H)-one;
1-ethyl-7-(1H-indazol-4-yl)-3,4-dihydropyrazino[2,3-b]pyrazin-2(1H)-one;
7-(pyridin-4-yl)-1-(2-(tetrahydro-2H-pyran-4-yl)ethyl)-3,4-dihydropyrazino[2,3-b]pyrazin- 2(1H)-one;
7-(6-aminopyridin-3-yl)-1-(2-(tetrahydro-2H-pyran-4-yl)ethyl)-3,4-dihydropyrazino[2,3- b]pyrazin-2(1H)-one;
1-methyl-7-(2-methyl-6-(4H-1,2,4-triazol-3-yl)pyridin-3-yl)-3,4-dihydropyrazino[2,3-b]pyrazin- 2(1H)-one;
2-(2-hydroxypropan-2-yl)-5-(8-(trans-4-methoxycyclohexyl)-7-oxo-5,6,7,8- tetrahydropyrazino[2,3-b]pyrazin-2-yl)pyridine 1-oxide;
4-methyl-5-(7-oxo-8-((tetrahydro-2H-pyran-4-yl)methyl)-5,6,7,8-tetrahydropyrazino[2,3- b]pyrazin-2-yl)picolinamide;
5-(8-((cis-4-methoxycyclohexyl)methyl)-7-oxo-5,6,7,8-tetrahydropyrazino[2,3-b]pyrazin-2-yl)- 4-methylpicolinamide;
7-(1H-pyrazol-4-yl)-1-(2-(tetrahydro-2H-pyran-4-yl)ethyl)-3,4-dihydropyrazino[2,3-b]pyrazin- 2(1H)-one;
1-(trans-4-methoxycyclohexyl)-7-(4-methyl-6-(1H-1,2,4-triazol-3-yl)pyridin-3-yl)-3,4- dihydropyrazino[2,3-b]pyrazin-2(1H)-one;
3-((7-(2-methyl-6-(4H-1,2,4-triazol-3-yl)pyridin-3-yl)-2-oxo-3,4-dihydropyrazino[2,3-b]pyrazin- 1(2H)-yl)methyl)benzonitrile; 1-((trans-4-methoxycyclohexyl)methyl)-7-(4-methyl-6-(1H-1,2,4-triazol-3-yl)pyridin-3-yl)-3,4- dihydropyrazino[2,3-b]pyrazin-2(1H)-one;
3-(7-oxo-8-(2-(tetrahydro-2H-pyran-4-yl)ethyl)-5,6,7,8-tetrahydropyrazino[2,3-b]pyrazin-2- yl)benzamide;
5-(8-((trans-4-methoxycyclohexyl)methyl)-7-oxo-5,6,7,8-tetrahydropyrazino[2,3-b]pyrazin-2- yl)-4-methylpicolinamide;
3-((7-(6-(2-hydroxypropan-2-yl)pyridin-3-yl)-2-oxo-3,4-dihydropyrazino[2,3-b]pyrazin-1(2H)- yl)methyl)benzonitrile;
7-(6-(2-hydroxypropan-2-yl)pyridin-3-yl)-1-((1R,3R)-3-methoxycyclopentyl)-3,4- dihydropyrazino[2,3-b]pyrazin-2(1H)-one;
7-(6-(2-hydroxypropan-2-yl)pyridin-3-yl)-1-((1S,3R)-3-methoxycyclopentyl)-3,4- dihydropyrazino[2,3-b]pyrazin-2(1H)-one;
7-(6-(2-hydroxypropan-2-yl)pyridin-3-yl)-1-((1S,3S)-3-methoxycyclopentyl)-3,4- dihydropyrazino[2,3-b]pyrazin-2(1H)-one;
7-(6-(2-hydroxypropan-2-yl)pyridin-3-yl)-1-((1R,3S)-3-methoxycyclopentyl)-3,4- dihydropyrazino[2,3-b]pyrazin-2(1H)-one;
7-(1H-indazol-6-yl)-1-(2-(tetrahydro-2H-pyran-4-yl)ethyl)-3,4-dihydropyrazino[2,3-b]pyrazin- 2(1H)-one;
7-(2-methyl-6-(4H-1,2,4-triazol-3-yl)pyridin-3-yl)-1-(2-morpholinoethyl)-3,4- dihydropyrazino[2,3-b]pyrazin-2(1H)-one;
1-(trans-4-hydroxycyclohexyl)-7-(2-methyl-6-(4H-1,2,4-triazol-3-yl)pyridin-3-yl)-3,4- dihydropyrazino[2,3-b]pyrazin-2(1H)-one;
1-(cis-4-hydroxycyclohexyl)-7-(2-methyl-6-(4H-1,2,4-triazol-3-yl)pyridin-3-yl)-3,4- dihydropyrazino[2,3-b]pyrazin-2(1H)-one;
7-(6-(2-hydroxypropan-2-yl)pyridin-3-yl)-1-(2-morpholinoethyl)-3,4-dihydropyrazino[2,3- b]pyrazin-2(1H)-one;
1-isopropyl-7-(2-methyl-6-(4H-1,2,4-triazol-3-yl)pyridin-3-yl)-3,4-dihydropyrazino[2,3- b]pyrazin-2(1H)-one;
7-(1H-imidazo[4,5-b]pyridin-6-yl)-1-(2-(tetrahydro-2H-pyran-4-yl)ethyl)-3,4- dihydropyrazino[2,3-b]pyrazin-2(1H)-one; 1-((cis-4-methoxycyclohexyl)methyl)-7-(2-methyl-6-(1H-1,2,4-triazol-3-yl)pyridin-3-yl)-3,4- dihydropyrazino[2,3-b]pyrazin-2(1H)-one;
1-(trans-4-hydroxycyclohexyl)-7-(6-(2-hydroxypropan-2-yl)pyridin-3-yl)-3,4- dihydropyrazino[2,3-b]pyrazin-2(1H)-one;
1-(cis-4-hydroxycyclohexyl)-7-(6-(2-hydroxypropan-2-yl)pyridin-3-yl)-3,4-dihydropyrazino[2,3- b]pyrazin-2(1H)-one;
4-(7-oxo-8-(2-(tetrahydro-2H-pyran-4-yl)ethyl)-5,6,7,8-tetrahydropyrazino[2,3-b]pyrazin-2- yl)benzamide;
7-(1H-indazol-5-yl)-1-(2-(tetrahydro-2H-pyran-4-yl)ethyl)-3,4-dihydropyrazino[2,3-b]pyrazin- 2(1H)-one;
7-(1H-pyrrolo[2,3-b]pyridin-5-yl)-1-(2-(tetrahydro-2H-pyran-4-yl)ethyl)-3,4- dihydropyrazino[2,3-b]pyrazin-2(1H)-one;
7-(2-methyl-6-(4H-1,2,4-triazol-3-yl)pyridin-3-yl)-1-(tetrahydro-2H-pyran-4-yl)-3,4- dihydropyrazino[2,3-b]pyrazin-2(1H)-one;
1-((1S,3R)-3-methoxycyclopentyl)-7-(2-methyl-6-(4H-1,2,4-triazol-3-yl)pyridin-3-yl)-3,4- dihydropyrazino[2,3-b]pyrazin-2(1H)-one;
1-((1R,3R)-3-methoxycyclopentyl)-7-(2-methyl-6-(4H-1,2,4-triazol-3-yl)pyridin-3-yl)-3,4- dihydropyrazino[2,3-b]pyrazin-2(1H)-one;
1-((1R,3S)-3-methoxycyclopentyl)-7-(2-methyl-6-(4H-1,2,4-triazol-3-yl)pyridin-3-yl)-3,4- dihydropyrazino[2,3-b]pyrazin-2(1H)-one;
1-((1S,3S)-3-methoxycyclopentyl)-7-(2-methyl-6-(4H-1,2,4-triazol-3-yl)pyridin-3-yl)-3,4- dihydropyrazino[2,3-b]pyrazin-2(1H)-one;
7-(1H-indol-5-yl)-1-(2-(tetrahydro-2H-pyran-4-yl)ethyl)-3,4-dihydropyrazino[2,3-b]pyrazin- 2(1H)-one;
1-ethyl-7-(2-methyl-6-(4H-1,2,4-triazol-3-yl)pyridin-3-yl)-3,4-dihydropyrazino[2,3-b]pyrazin- 2(1H)-one;
7-(1H-indol-6-yl)-1-(2-(tetrahydro-2H-pyran-4-yl)ethyl)-3,4-dihydropyrazino[2,3-b]pyrazin- 2(1H)-one;
7-(4-(2-hydroxypropan-2-yl)phenyl)-1-(trans-4-methoxycyclohexyl)-3,4-dihydropyrazino[2,3- b]pyrazin-2(1H)-one; 7-(6-(2-hydroxypropan-2-yl)pyridin-3-yl)-1-(tetrahydro-2H-pyran-4-yl)-3,4- dihydropyrazino[2,3-b]pyrazin-2(1H)-one;
1-((trans-4-methoxycyclohexyl)methyl)-7-(2-methyl-6-(1H-1,2,4-triazol-3-yl)pyridin-3-yl)-3,4- dihydropyrazino[2,3-b]pyrazin-2(1H)-one;
7-(6-(2-hydroxypropan-2-yl)pyridin-3-yl)-1-((cis-4-methoxycyclohexyl)methyl)-3,4- dihydropyrazino[2,3-b]pyrazin-2(1H)-one;
1-(2-methoxyethyl)-7-(4-methyl-2-(methylamino)-1H-benzo[d]imidazol-6-yl)-3,4- dihydropyrazino[2,3-b]pyrazin-2(1H)-one;
7-(7-methyl-2-oxo-2,3-dihydro-1H-benzo[d]imidazol-5-yl)-1-((tetrahydro-2H-pyran-4- yl)methyl)-3,4-dihydropyrazino[2,3-b]pyrazin-2(1H)-one;
7-(2-methyl-4-(4H-1,2,4-triazol-3-yl)phenyl)-3,4-dihydropyrazino[2,3-b]pyrazin-2(1H)-one; 1-(2-methoxyethyl)-7-(4-methyl-6-(1H-1,2,4-triazol-3-yl)pyridin-3-yl)-3,4-dihydropyrazino[2,3- b]pyrazin-2(1H)-one;
1-benzyl-7-(2-methyl-4-(4H-1,2,4-triazol-3-yl)phenyl)-3,4-dihydropyrazino[2,3-b]pyrazin- 2(1H)-one;
7-(3-fluoro-4-(4H-1,2,4-triazol-3-yl)phenyl)-1-(2-methoxyethyl)-3,4-dihydropyrazino[2,3- b]pyrazin-2(1H)-one;
7-(3-fluoro-4-(4H-1,2,4-triazol-3-yl)phenyl)-1-(2-(tetrahydro-2H-pyran-4-yl)ethyl)-3,4- dihydropyrazino[2,3-b]pyrazin-2(1H)-one;
7-(3-fluoro-2-methyl-4-(1H-1,2,4-triazol-3-yl)phenyl)-1-(2-methoxyethyl)-3,4- dihydropyrazino[2,3-b]pyrazin-2(1H)-one;
1-(trans-4-methoxycyclohexyl)-7-(2-methyl-6-(4H-1 ,2,4-triazol-3-yl)pyridin-3-yl)-3,4- dihydropyrazino[2,3-b]pyrazin-2(1H)-one;
7-(6-(2-hydroxypropan-2-yl)pyridin-3-yl)-1-(trans-4-methoxycyclohexyl)-3,4- dihydropyrazino[2,3-b]pyrazin-2(1H)-one;
7-(5-fluoro-2-methyl-4-(4H-1,2,4-triazol-3-yl)phenyl)-1-(2-(tetrahydro-2H-pyran-4-yl)ethyl)- 3,4-dihydropyrazino[2,3-b]pyrazin-2(1H)-one;
7-(3-fluoro-2-methyl-4-(1H-1,2,4-triazol-3-yl)phenyl)-1-(2-(tetrahydro-2H-pyran-4-yl)ethyl)- 3,4-dihydropyrazino[2,3-b]pyrazin-2(1H)-one; 1-(2-methoxyethyl)-7-(2-methyl-6-(4H-1,2,4-triazol-3-yl)pyridin-3-yl)-3,4-dihydropyrazino[2,3- b]pyrazin-2(1H)-one;
7-(6-(2-hydroxypropan-2-yl)pyridin-3-yl)-1-((trans-4-methoxycyclohexyl)methyl)-3,4- dihydropyrazino[2,3-b]pyrazin-2(1H)-one;
1-(cyclopentylmethyl)-7-(6-(2-hydroxypropan-2-yl)pyridin-3-yl)-3,4-dihydropyrazino[2,3- b]pyrazin-2(1H)-one;
7-(4-(2-hydroxypropan-2-yl)phenyl)-1-(2-methoxyethyl)-3,4-dihydropyrazino[2,3-b]pyrazin- 2(1H)-one;
(S)-7-(6-(1-hydroxyethyl)pyridin-3-yl)-1-(2-(tetrahydro-2H-pyran-4-yl)ethyl)-3,4- dihydropyrazino[2,3-b]pyrazin-2(1H)-one;
(R)-7-(6-(1-hydroxyethyl)pyridin-3-yl)-1-(2-(tetrahydro-2H-pyran-4-yl)ethyl)-3,4- dihydropyrazino[2,3-b]pyrazin-2(1H)-one;
7-(2-methyl-6-(4H-1,2,4-triazol-3-yl)pyridin-3-yl)-1-((tetrahydro-2H-pyran-4-yl)methyl)-3,4- dihydropyrazino[2,3-b]pyrazin-2(1H)-one;
7-(4-(2-hydroxypropan-2-yl)phenyl)-1-(2-(tetrahydro-2H-pyran-4-yl)ethyl)-3,4- dihydropyrazino[2,3-b]pyrazin-2(1H)-one;
7-(6-(2-hydroxypropan-2-yl)pyridin-3-yl)-1-(4-(trifluoromethyl)benzyl)-3,4- dihydropyrazino[2,3-b]pyrazin-2(1H)-one;
7-(6-(2-hydroxypropan-2-yl)pyridin-3-yl)-1-(3-(trifluoromethyl)benzyl)-3,4- dihydropyrazino[2,3-b]pyrazin-2(1H)-one;
7-(6-(2-hydroxypropan-2-yl)pyridin-3-yl)-1-(3-methoxypropyl)-3,4-dihydropyrazino[2,3- b]pyrazin-2(1H)-one;
7-(4-methyl-6-(1H-1,2,4-triazol-3-yl)pyridin-3-yl)-1-(2-(tetrahydro-2H-pyran-4-yl)ethyl)-3,4- dihydropyrazino[2,3-b]pyrazin-2(1H)-one;
7-(6-(2-hydroxypropan-2-yl)pyridin-3-yl)-1-(2-methoxyethyl)-3,4-dihydropyrazino[2,3- b]pyrazin-2(1H)-one;
7-(6-(2-hydroxypropan-2-yl)pyridin-3-yl)-1-((tetrahydro-2H-pyran-4-yl)methyl)-3,4- dihydropyrazino[2,3-b]pyrazin-2(1H)-one;
7-(4-methyl-2-(methylamino)-1H-benzo[d]imidazol-6-yl)-1-((tetrahydro-2H-pyran-4-yl)methyl)- 3,4-dihydropyrazino[2,3-b]pyrazin-2(1H)-one; 7-(2-amino-4-methyl-1H-benzo[d]imidazol-6-yl)-1-((tetrahydro-2H-pyran-4-yl)methyl)-3,4- dihydropyrazino[2,3-b]pyrazin-2(1H)-one;
7-(2-methyl-6-(4H-1,2,4-triazol-3-yl)pyridin-3-yl)-1-(2-(tetrahydro-2H-pyran-4-yl)ethyl)-3,4- dihydropyrazino[2,3-b]pyrazin-2(1H)-one;
(R)-7-(6-(2-hydroxypropan-2-yl)pyridin-3-yl)-3-methyl-1-(2-(tetrahydro-2H-pyran-4-yl)ethyl)- 3,4-dihydropyrazino[2,3-b]pyrazin-2(1H)-one;
(S)-7-(6-(2-hydroxypropan-2-yl)pyridin-3-yl)-3-methyl-1-(2-(tetrahydro-2H-pyran-4-yl)ethyl)- 3,4-dihydropyrazino[2,3-b]pyrazin-2(1H)-one;
7-(6-(2-hydroxypropan-2-yl)pyridin-3-yl)-3,3-dimethyl-1-(2-(tetrahydro-2H-pyran-4-yl)ethyl)- 3,4-dihydropyrazino[2,3-b]pyrazin-2(1H)-one;
7-(2-amino-4-methyl-1H-benzo[d]imidazol-6-yl)-1-(2-(tetrahydro-2H-pyran-4-yl)ethyl)-3,4- dihydropyrazino[2,3-b]pyrazin-2(1H)-one;
7-(6-(2-hydroxypropan-2-yl)pyridin-3-yl)-1-(2-(tetrahydro-2H-pyran-4-yl)ethyl)-3,4- dihydropyrazino[2,3-b]pyrazin-2(1H)-one;
7-(2-methyl-4-(1H-1,2,4-triazol-3-yl)phenyl)-1-(2-(tetrahydro-2H-pyran-4-yl)ethyl)-3,4- dihydropyrazino[2,3-b]pyrazin-2(1H)-one;
7-(4-(1H-1,2,4-triazol-5-yl)phenyl)-1-(2-(tetrahydro-2H-pyran-4-yl)ethyl)-3,4- dihydropyrazino[2,3-b]pyrazin-2(1H)-one;
1-(1-hydroxypropan-2-yl)-7-(2-methyl-6-(1H-1,2,4-triazol-3-yl)pyridin-3-yl)-3,4- dihydropyrazino[2,3-b]pyrazin-2(1H)-one; and
1-(2-hydroxyethyl)-7-(2-methyl-6-(1 H-1,2,4-triazol-3-yl)pyridin-3-yl)-3,4-dihydropyrazino[2,3- b]pyrazin-2(1H)-one, and pharmaceutically acceptable salts, clathrates, solvates, stereoisomers, tautomers, metabolites, isotopologues and prodrugs thereof.
[00132] In some embodiments, a TOR inhibitor of Formula (I) is 7-(6-(2-hydroxypropan-2- yl)pyridin-3-yl)-1-((1r,4r)-4-methoxycyclohexyl)-3,4-dihydropyrazino-[2,3-b]pyrazin-2(1H)- one, alternatively named 7-(6-(2-hydroxypropan-2-yl)pyridin-3-yl)-1-((trans)-4- methoxycyclohexyl)-3,4-dihydropyrazino[2,3-b]pyrazin-2(1H)-one, or 7-(6-(2-hydroxypropan- 2-yl)pyridin-3-yl)-1-((1R*,4R*)-4-methoxycyclohexyl)-3,4-dihydropyrazino[2,3-b]pyrazin- 2(1H)-one, depicted below and referred to herein as Compound 2:
Figure imgf000037_0001
[00133] In some embodiments, a TOR inhibitor of Formula (I) is a pharmaceutically acceptable salt of Compound 2.
[00134] In some embodiments, a TOR inhibitor of Formula (I) is 1 -((trans)-4- hydroxycyclohexyl)-7-(6-(2-hydroxypropan-2-yl)pyridin-3-yl)-3,4-dihydropyrazino[2,3- b]pyrazin-2(1H)-one, alternatively named 1-((1r,4r)-4-hydroxycyclohexyl)-7-(6-(2- hydroxypropan-2-yl)pyridin-3-yl)-3,4-dihydropyrazino[2,3-b]pyrazin-2(1H)-one, depicted below and referred to herein as Compound 3:
Figure imgf000037_0002
[00135] In some embodiments, a TOR inhibitor of Formula (I) is a pharmaceutically acceptable salt of Compound 3.
[00136] In some embodiments, a TOR inhibitor of Formula (I) is 1-ethyl-7-(2-methyl-6-(1H- 1,2,4-triazol-3-yl)pyridin-3-yl)-3,4-dihydropyrazino[2,3-b]pyrazin-2(1H)-one, or a tautomer thereof, for example, 1-ethyl-7-(2-methyl-6-(4H-1,2,4-triazol-3-yl)pyridin-3-yl)-3,4- dihydropyrazino[2,3-b]pyrazin-2(1H)-one, or 1-ethyl-7-(2-methyl-6-(1H-1,2,4-triazol-5- yl)pyridin-3-yl)-3,4-dihydropyrazino[2,3-b]pyrazin-2(1H)-one. One such tautomer is depicted below and referred to herein as Compound 4:
Figure imgf000038_0001
[00137] In some embodiments, a TOR inhibitor of Formula (I) is a pharmaceutically acceptable salt of Compound 4.
[00138] TOR inhibitors can be prepared via standard, well-known synthetic methodology, see e.g., March, J. Advanced Organic Chemistry; Reactions Mechanisms, and Structure, 4th ed., 1992. Starting materials useful for preparing compounds of formula (I) and intermediates therefore, are commercially available or can be prepared from commercially available materials using known synthetic methods and reagents.
[00139] Particular methods for preparing compounds of formula (I) are disclosed in U.S. Patent No. 8,110,578, issued February 7, 2012, and U.S. Publication No. 2011/0137028, filed October 25, 2010, incorporated by reference herein in their entirety.
[00140] In some embodiments, a TOR inhibitor is selected from rapamycin or an analog thereof. For instance, in some embodiments, a rapamycin analog in the form of an ester, ether, hydrazone, hydroxylamine, or oxime derivatives of rapamycin. In some embodiments, a TOR inhibitor is AP23573 (Deforolimus), AP-23675, AP-23841, ABT-578 (Zotarolimus), CCI779 (Temsirolimus), RAD-001 (Everolimus), 7-epi-rapamycin, 7-thiomethyl-rapamycin, 7-epi- trimethoxyphenyl-rapamycin, 7-epi-thiomethyl-rapamycin, and 7-demethoxy-rapamycin, 32- demethoxy-rapamycin, 2-desmethyl-rapamycin, 42-O-(2-hydroxy)ethyl rapamycin, Ridaforolimus, AZD8055, OSI-027, INK-128 (MLN0128), wortmannin, and LY29004.
[00141] In some embodiments, a TOR inhibitor is selected from Everolimus, Temsirolimus, Ridaforolimus, AZD8055, OSI-027, INK-128 (MLN0128), wortmannin, and LY29004.
[00142] In some embodiments, a TOR inhibitor is a compound having the following structure:
Figure imgf000039_0001
Figure imgf000039_0002
or a pharmaceutically acceptable salt, clathrate, solvate, stereoisomer, tautomer, or prodrug thereof.
[00144] In some embodiments, a TOR inhibitor is a compound having the following structure:
Figure imgf000040_0001
or a pharmaceutically acceptable salt, clathrate, solvate, stereoisomer, tautomer, or prodrug thereof.
Figure imgf000040_0002
[00145] In some embodiments, a TOR inhibitor is a compound having the following structure:
Figure imgf000040_0003
or a pharmaceutically acceptable salt, clathrate, solvate, stereoisomer, tautomer, or prodrug thereof.
[00146] In some embodiments, a TOR inhibitor is a compound having the following structure:
Figure imgf000041_0001
clathrate,
[00147] In some embodiments, a TOR inhibitor is a compound having the following structure:
Figure imgf000041_0002
or a pharmaceutically acceptable salt, clathrate, solvate, stereoisomer, tautomer, or prodrug thereof.
[00148] In some embodiments, a TOR inhibitor is a compound having the following structure:
Figure imgf000042_0001
or a pharmaceutically a utomer, or prodrug thereof.
[00149] In some embodiments, a TOR inhibitor is a compound having the following structure:
Figure imgf000042_0002
or a pharmaceutically acceptable salt, clathrate, solvate, stereoisomer, tautomer, or prodrug thereof.
[00150] In some embodiments, a TOR inhibitor is a compound having the following structure:
Figure imgf000042_0003
or a pharmaceutically acceptable salt, clathrate, solvate, stereoisomer, tautomer, or prodrug thereof.
[00151] In some embodiments, a TOR inhibitor is a compound having the following structure:
Figure imgf000043_0001
or a pharmaceutically acceptable salt, clathrate, solvate, stereoisomer, tautomer, or prodrug thereof. [00152] In some embodiments, a TOR inhibitor is a compound disclosed in the patent publications listed below in Table B.
Table B
[00153] WO 2008/023161 (see, e.g., page 5, line 5 to page 11, line 15), WO 2009/007751 (see, e.g., page 9, line 8 to page 26, line 8), WO 2009/007749 (see, e.g., page 9, line 21 to page 29, line 23), WO 2009/007750 (see, e.g., page 9, line 21 to page 32, line 22), WO 2009/007748 (see, e.g., page 9, line 6 to page 42, line 28), WO 2008/032028 (see, e.g., page 11, line 13 to page 21, line 13), WO 2008/032086 (see, e.g., page 10 line 21 to page 15, line 22), WO 2008/032072 (see, e.g., page 11, line 11 to page 16, line 13), WO 2008/032033 (see, e.g., page 11, line 3 to page 16, line 5), WO 2008/032089 (see, e.g., page 11, line 11 to page 16, line 13), WO 2008/032060 (see, e.g., page 11, line 3 to page page 16, line 6), WO 2008/032091 (see, e.g., page 11, line 11 to page 16, line 13), WO 2008/032036 (see, e.g., page 11, line 13 to page 21, line 13), WO 2008/032077 (see, e.g., page 10, line 21 to page 15, line 22), WO 2008/032064 (see, e.g., page 11, line 3 to page 16, line 5), WO 2008/032027 (see, e.g., page 10, line 21 to page 15, line 22), WO 2007/135398 (see, e.g., page 11, line 28 to page 16, line 6), WO 2007/129052 (see, e.g., page 10, line 8 to page 13, line 5), WO 2007/129044 (see, e.g., page 10, line 22 to page 13, line 20), WO 2007/080382 (see, e.g., page 9, line 20 to page 32, line 32), WO 2007/066102 (see, e.g., page 9, line 22 to page 14, line 17), WO 2007/066099 (see, e.g., page 9, line 22 to page 14, line 14), WO 2007/066103 (see, e.g., page 9, line 22 to page 14, line 16), WO 2007/060404 (see, e.g., 5, line 4 to page 7, line 25), WO 2006/090169 (see, e.g., page 4, lines 1-25), WO 2006/090167 (see, e.g., page 3, line 33 to page 6, line 23), WO 2008/115974 (see, e.g., page 4, paragraph [0012] to page 127, paragraph [0257]), WO 2009/052145 (see, e.g., page 5, paragraph [0015] to page 81, paragraph [0082]), WO 2010/006072 (see, e.g., page 28, line 1 to page 34, line 1), WO 2007/044698 (see, e.g., page 3, paragraph [0010] to the bottom of page 7), WO 2007/044813 (see, e.g., page 3, paragraph [0010] to the middle of page 7), WO 2007/044729 (see, e.g., page 3, paragraph [0010] to the bottom of page 10), WO 2007/129161 (see, e.g., page 2, line 10 to page 9, line 19), WO 2006/046031 (see, e.g., page 2, line 15 to page 4, line 12), WO 2003/072557 (see, e.g., page 1, line 4 to page 2, line 27), WO 2004/048365 (see, e.g., page 1, line 4 to page 4, line 17), WO 2004/078754 (see, e.g., page 1, line 4 to page 2, line 21), WO 2004/096797 (see, e.g., page 1, line 4 to page 2, line 34), WO 2005/021519 (see, e.g., page 1, line 4 to page 4, line 17) or US 2007/112005 (see, e.g., page 2, paragraph [0012] to page 22, paragraph [0065]), each of which is incorporated by reference herein in its entirety. Methods of Treatment
[00154] As described generally above, Compound 1, and pharmaceutically acceptable salts thereof described herein, is an inhibitor of one or both of ERK1 and ERK2. One of ordinary skill in the art will recognize that ERK is one of the key components in the RAS-RAF-MEK-ERK MAPK pathway and that ERK1 and ERK2 are downstream nodes within the MAPK pathway. Without wishing to be bound by theory, because of the downstream location of ERK1 and ERK1 in the MAPK pathway, an ERK inhibitor can treat disease or disorders in which activation of the MAPK pathway at any level (Ras-Raf-Mek-ERK) is known or suspected to play a role, including one or both of ERK1 and ERK2 as well as other nodes in the MAPK pathway upstream from ERK (such as Ras, Raf and Mek). Furthermore, because ERK is a downstream target, ERK inhibitors are believed to be able to overcome, in some instances, drug resistance induced by inhibitors of targets upstream of ERK within the MAPK pathway. For example, small molecule inhibitors of RAF or MEK utilized in the treatment of K-RAS and B-RAF mutant tumors have resulted in such drug resistance. Similarly, drug resistance has been associated with other tumors driven by hyperactivation of the MAPK pathway (such as NF1 mutant tumors). Kinase selectivity was achieved through silencing the selective Cys in a combination of the interactions between the covalent inhibitors of the invention and unique amino acids in the ATP binding pocket. Targeting the selective Cys provides for prolonged pharmacodynamics in silencing ERK activity, as well as potential lower doses in cancer treatment, compared to reversible inhibitors. [00155] The activity of Compound 1, and pharmaceutically acceptable salts thereof, as an inhibitor of one or both of an ERK1 and ERK2 kinase, or a mutant thereof, may be assayed in vitro, in vivo or in a cell line. In vitro assays include assays that determine inhibition of downstream phosphorylation, changes in gene expression, subsequent functional markers and consequences, and/or kinase activity of one or both of activated ERK1 and ERK2 kinase, or a mutant thereof. Alternate in vitro assays quantitate the ability of the test compound to bind to one or both of ERK1 and ERK2. Test compound binding may be measured by radiolabeling the test compound prior to binding, isolating one or both of the compound / ERK1 complex and the compound / ERK2 complex, and determining the amount of radiolabel bound. Alternatively, test compound binding may be determined by running a competition experiment where test compounds are incubated with one or both of ERK1 and ERK2 kinase bound to known radioligands. Test compound binding may be determined by competition with an ERK covalent probe that is amenable to further functionalization with a detection probe, such as, for example, a fluorophore, biotin conjugate, radiolabel, or any other probe that facilitates its quantification. Detailed conditions for assaying a compound utilized in this invention as an inhibitor of one or both of ERK1 and ERK2, or a mutant thereof, are also set forth below and/or in the Examples of the‘230 publication.
[00156] The term "measurably inhibit", as used herein means a measurable change in one or both of ERK1 and ERK2 protein kinase activity between a sample comprising a provided composition, and one or both of an ERK1 and ERK2 protein kinase and an equivalent sample comprising one or both of ERK1 and ERK2 protein kinase in the absence of a provided composition. Such measurements of protein kinase activity are known to one of ordinary skill in the art and include those methods set forth herein below and/or in the Examples of the‘230 publication.
[00157] As described above, in some embodiments, Compound 1, and pharmaceutically acceptable salts thereof, either alone or in combination with another agent such as a TOR inhibitor, are inhibitors of one or both of ERK1 and ERK2 protein kinases, and ERK1 and ERK2 are downstream targets within the MAPK pathway. Without wishing to be bound by any particular theory, such compounds and compositions are particularly useful for treating or lessening the severity of a disease, condition, or disorder in which activation of the MAPK pathway at any level (Ras-Raf-Mek-ERK) is known or suspected to play a role. Such disease, condition, or disorder may be referred to herein as associated with the MAPK pathway or alternatively as associated with one or both of ERK1 and ERK2. Such diseases, conditions, or disorders may also be refererd to herein as an "ERK1- or ERK2-mediated disease, condition, or disorder."
[00158] In some embodiments, the present invention provides a method for treating or lessening the severity of a disease, condition, or disorder where activation of the MAPK pathway (at any level in Ras-Raf-Mek-ERK), including one or both of ERK1 and ERK2 protein kinases, is implicated in said disease, condition, or disorder wherein said method comprises administering to a patient in need thereof Compound 1, or a pharmaceutically acceptable salt thereof, in combination with a TOR inhibitor.
[00159] In some embodiments, the present invention relates to a method of inhibiting one or both of ERK1 and ERK2 protein kinase activity in a patient comprising the step of administering to said patient a composition comprising Compound 1, or a pharmaceutically acceptable salt thereof, in combination with a TOR inhibitor, or a composition comprising any of the foregoing.
[00160] In other embodiments, the present invention provides a method for treating a disease, condition, or disorder mediated by one or both of ERK1 and ERK2 kinase, or a mutant thereof, in a patient in need thereof, comprising the step of administering to said patient Compound 1, or pharmaceutically acceptable salts thereof, in combination with a TOR inhibitor, or a pharmaceutically acceptable composition comprising any of the foregoing. Such disorders are described in detail herein.
[00161] In certain embodiments, the present invention provides a method for overcoming drug resistance to Raf or MEK inhibitors, comprising the step of administering to a patient an inhibitor compound of one or both of ERK1 and ERK2, such as Compound 1, or a pharmaceutically acceptable salt thereof, either alone or in combination with a Tor inhibitor. In certain embodiments, the mechanism of drug resistance is through mutation of a target protein or reactivation of the MAPK pathway.
[00162] As used herein, the term“resistance” may refer to changes in a wild-type nucleic acid sequence coding a target protein, and/or to the amino acid sequence of the target protein and/or to the amino acid sequence of another protein, which changes, decreases or abolishes the inhibitory effect of the inhibitor on the target protein. The term“resistance” may also refer to overexpression or silencing of a protein differing from a target protein that can reactivate the MAPK pathway or other survival pathways.
[00163] In some embodiments, treatment is administered after one or more symptoms have developed. In other embodiments, treatment is administered in the absence of symptoms. For example, treatment is administered to a susceptible individual prior to the onset of symptoms (e.g., in light of a history of symptoms and/or in light of genetic or other susceptibility factors). Treatment is also continued after symptoms have resolved, for example to prevent, delay or lessen the severity of their recurrence.
[00164] In some embodiments, the present invention provides a method of treating, stabilizing or lessening the severity or progression of one or more diseases or disorders associated with one or both of ERK1 and ERK2 comprising administering to a patient in need thereof a composition comprising Compound 1, or a pharmaceutically acceptable salt thereof, in combination with a TOR inhibitor of Formula (I).
[00165] In some embodiments, the present invention provides a method of treating, stabilizing or lessening the severity or progression of one or more diseases or disorders associated with one or both of ERK1 and ERK2 comprising administering to a patient in need thereof a composition comprising Compound 1, or a pharmaceutically acceptable salt thereof, in combination with a compound selected from Compound 2, Compound 3, or Compound 4, or a pharmaceutically acceptable salt thereof.
[00166] In some embodiments, the present invention provides a method of treating, stabilizing or lessening the severity or progression of one or more diseases or disorders associated with one or both of ERK1 and ERK2 comprising administering to a patient in need thereof a composition comprising Compound 1, or a pharmaceutically acceptable salt thereof, in combination with Compound 2, or a pharmaceutically acceptable salt thereof.
[00167] In some embodiments, the present invention provides a method of treating, stabilizing or lessening the severity or progression of one or more diseases or disorders associated with one or both of ERK1 and ERK2 comprising administering to a patient in need thereof a composition comprising Compound 1, or a pharmaceutically acceptable salt thereof, in combination with Compound 3, or a pharmaceutically acceptable salt thereof. [00168] In some embodiments, the present invention provides a method of treating, stabilizing or lessening the severity or progression of one or more diseases or disorders associated with one or both of ERK1 and ERK2 comprising administering to a patient in need thereof a composition comprising Compound 1, or a pharmaceutically acceptable salt thereof, in combination with Compound 4, or a pharmaceutically acceptable salt thereof.
[00169] In some embodiments, the present invention provides a system for treating, stabilizing or lessening the severity of one or more diseases or disorders associated with one or more of ERK1 and ERK2, the system comprising Compound 1, or a pharmaceutically acceptable salt thereof, in combination with a TOR inhibitor. In some such embodiments, the present invention contemplates a system comprising any of the above-described TOR inhibitors.
[00170] In some embodiments, a TOR inhibitor is selected from rapamycin, AP23573 (Deforolimus), AP-23675, AP-23841, ABT-578 (Zotarolimus), CCI779 (Temsirolimus), RAD- 001 (Everolimus), 7-epi-rapamycin, 7-thiomethyl-rapamycin, 7-epi-trimethoxyphenyl- rapamycin, 7-epi-thiomethyl-rapamycin, and 7-demethoxy-rapamycin, 32-demethoxy- rapamycin, 2-desmethyl-rapamycin, 42-O-(2-hydroxy)ethyl rapamycin, Ridaforolimus, AZD8055, OSI-027, INK-128, MLN0128, wortmannin, and LY29004.
[00171] In some embodiments, the present invention provides a system for treating, stabilizing or lessening the severity of one or more diseases or disorders associated with one or more of ERK1 and ERK2, the system comprising Compound 1, or a pharmaceutically acceptable salt thereof, in combination with a TOR inhibitor, wherein the TOR inhibitor is a compound of Formula (I). For instance, in some embodiments, a TOR inhibitor is selected from any of Compound 2, Compound 3, or Compound 4, or a pharmaceutically acceptable salt thereof.
[00172] General diseases, conditions, or disorders treated by Compound 1, and pharmaceutically acceptable salts thereof, in combination with a TOR inhibitor include cancer, an autoimmune disorder, a neurodegenerative or neurological disorder, liver disease, a cardiac disorder, schizophrenia, or a bone-related disorder.
[00173] In some embodiments, the present invention provides a method for treating an ERK1- or ERK2-mediated disease, condition, or disorder comprising administering to a patient in need thereof Compound 1, or a pharmaceutically acceptable salt thereof, in combination with a TOR inhibitor. [00174] In some embodiments, the present invention relates to a method of treating or lessening the severity of a disease, condition, or disorder selected from cancer, stroke, diabetes, hepatomegaly, cardiovascular disease including cardiomegaly, Alzheimer's disease, cystic fibrosis, viral disease, autoimmune diseases, atherosclerosis, restenosis, psoriasis, allergic disorders including asthma, inflammation, neurological disorders and hormone-related diseases, wherein the method comprises administering to a patient in need thereof a composition comprising Compound 1, or a pharmaceutically acceptable salt thereof, in combination with a TOR inhibitor.
[00175] In some embodiments, the present invention relates to a method of treating a cancer wherein the method comprises administering to a patient in need thereof a composition comprising Compound 1, or a pharmaceutically acceptable salt thereof, in combination with a TOR inhibitor. In some embodiments, the cancer is recurring. In certain embodiments, the cancer is refractory. In some embodiments, the cancer is metastatic. In some embodiments, the cancer is locally advanced.
[00176] In certain embodiments, the cancer is a RAF inhibitor-resistant cancer. In some such embodiments, the RAF inhibitor-resistant cancer is a BRAF inhibitor-resistant cancer.
[00177] In certain embodiments, the cancer is a MEK inhibitor-resistant cancer.
[00178] In certain embodiments, the cancer is a MAPK pathway-mediated cancer.
[00179] In some embodiments, the cancer is a BRAF-mutated cancer. In certain embodiments, the BRAF-mutated cancer is a BRAFV600-mutated cancer, such as BRAFV600E BRAFV600K, BRAFV600R, and BRAFV600D.
[00180] In some embodiments, the cancer is a RAS-mutated cancer. In certain embodiments, the RAS-mutated involves codons 12, 13, or 61. In certain embodiments, the RAS-mutated cancer is a KRAS-mutated cancer, including, but not limited to, KRASG12C/D/V, KRASG13C/D,or KRASQ61L/H/R. In certain embodiments, the RAS-mutated cancer is an NRAS-mutated cancer, including, but not limited to, NRASQ61R, NRASQ61K, NRASQ61L, or NRASQ61H. In certain embodiments, the RAS-mutated cancer is an HRAS-mutated cancer, including, but not limited to, HRASG12V, HRASQ61R, and HRASG12S.
[00181] In some embodiments, the present invention relates to a method of treating a cancer wherein the method comprises administering to a patient in need thereof a composition comprising Compound 1, or a pharmaceutically acceptable salt thereof, in combination with a TOR inhibitor, wherein the cancer is selected from multiple myeloma, breast, ovary, cervix, prostate, testis, genitourinary tract, esophagus, larynx, glioblastoma, neuroblastoma, stomach (gastric), skin, keratoacanthoma, lung, epidermoid carcinoma, large cell carcinoma, small cell carcinoma, lung, bone, colon, thyroid, adenoma, pancreas, adenocarcinoma, thyroid, follicular carcinoma, undifferentiated carcinoma, papillary carcinoma, seminoma, melanoma (including uveal melanoma) sarcoma, bladder carcinoma, liver carcinoma (e.g., hepatocellular carcinoma (HCC)) and biliary passage carcinoma), kidney carcinoma, myeloid disorders, lymphoid disorders, Hodgkin's, hairy cells, buccal cavity and pharynx (oral), lip, tongue, mouth, pharynx, small intestine, colorectal carcinoma, large intestine, rectum, brain and central nervous system, endometrial, multiple myeloma (MM), prostate, AML, and leukemia. In some such embodiments, the cancer is relapsed. In some embodiments, the cancer is refractory. In some embodiments, the cancer is locally advanced. In some embodiments, the cancer is metastatic.
[00182] In some embodiments, the present invention relates to a method of treating a cancer wherein the method comprises administering to a patient in need thereof a composition comprising Compound 1, or a pharmaceutically acceptable salt thereof, in combination with a TOR inhibitor, wherein the cancer is selected from carcinoma, lymphoma, blastoma, sarcoma, and leukemia. In some embodiments, a sarcoma is a soft tissue sarcoma. In some embodiments, a lymphoma is non-hodgkins lymphoma. In some embodiments, a lymphoma is large cell immunoblastic lymphoma. In some embodiments, the cancer is selected from adenocarcinoma; adenoma; adrenocortical cancer; bladder cancer; bone cancer; brain cancer; breast cancer; cancer of the buccal cavity; cervical cancer; colon cancer; colorectal cancer; endometrial or uterine carcinoma; epidermoid carcinoma; esophogeal cancer; eye cancer; follicular carcinoma; gallbladder cancer; prostate, AML, multiple myeloma (MM), gastrointestinal cancer, such as, for example, gastrointestinal stromal tumor; cancer of the genitourinary tract; glioblastoma; hairy cell carcinoma; various types of head and neck cancer; hepatic carcinoma; hepatocellular cancer; Hodgkin's disease; keratoacanthoma; kidney cancer; large cell carcinoma; cancer of the large intestine; laryngeal cancer; liver cancer; lung cancer, such as, for example, adenocarcinoma of the lung, anaplastic carcinoma of the lung, papillary lung adenocarcinoma, small-cell lung cancer, squamous carcinoma of the lung, non-small cell lung cancer; melanoma and nonmelanoma skin cancer; lymphoid disorders; myeloproliferative disorders, such as, for example, polycythemia vera, essential thrombocythemia, chronic idiopathic myelofibrosis, myeloid metaplasia with myelofibrosis, chronic myeloid leukemia (CML), chronic myelomonocytic leukemia, chronic eosinophilic leukemia, chronic lymphocytic leukemia (CLL), hypereosinophilic syndrome, systematic mast cell disease, atypical CML, AML, or juvenile myelomonocytic leukemia; plasmacytoma; multiple myeloma; neuroblastoma; ovarian cancer; papillary carcinoma; pancreatic cancer; cancer of the peritoneum; prostate cancer, including benign prostatic hyperplasia; rectal cancer; salivary gland carcinoma; sarcoma; seminoma; squamous cell cancer; small cell carcinoma; cancer of the small intestine; stomach cancer; testicular cancer; thyroid cancer; undifferentiated carcinoma; and vulval cancer. In some such embodiments, the cancer is relapsed. In some embodiments, the cancer is refractory. In some embodiments, the cancer is locally advanced. In some embodiments, the cancer is metastatic.
[00183] In some embodiments, the present invention relates to a method of treating a cancer wherein the method comprises administering to a patient in need thereof a composition comprising Compound 1, or a pharmaceutically acceptable salt thereof, in combination with a TOR inhibitor, wherein the cancer is selected from melanoma, pancreatic cancer, thyroid cancer, colorectal cancer, lung cancer (e.g., non-small cell lung cancer), breast cancer, endometrial cancer, prostate cancer, ovarian cancer, hepatocellular carcinoma (HCC), multiple myeloma (MM), and leukemia. In some embodiments, a leukemia is an acute leukemia. In certain embodiments, a leukemia is acute myeloid leukemia. In certain embodiments, a leukemia is acute lymphoblastic leukemia.
[00184] In some embodiments, the present invention relates to a method of treating a cancer wherein the method comprises administering to a patient in need thereof a composition comprising Compound 1, or a pharmaceutically acceptable salt thereof, in combination with a TOR inhibitor, wherein the cancer is selected from melanoma, colorectal cancer, lung cancer, or pancreatic.
[00185] In some embodiments, the present invention relates to a method of treating a cancer wherein the method comprises administering to a patient in need thereof a composition comprising Compound 1, or a pharmaceutically acceptable salt thereof, in combination with a TOR inhibitor, wherein the cancer is melanoma. In certain embodiments, the melanoma is uveal melanoma. In some embodiments, the melanoma is a melanoma of the skin. In certain embodiments, the melanoma is locally advanced. In some embodiments, the melanoma is metastatic. In some embodiments, the melanoma is recurring. In some embodiments, the melanoma is BRAFv600-mutated melanoma. In certain embodiments, the melanoma is a RAS- mutated melanoma. In some embodiments, the melanoma is NRAS-mutated melanoma. In certain embodiments, the melanoma is wild type for KRAS, NRAS or BRAF. In certain embodiments, the melanoma is a BRAF inhibitor-resistant (e.g., vemurfenib-resistant, dabrafenib-resistant, encorafenib-resistant, etc.) melanoma. In certain embodiments, the cancer is a VemR (i.e., Vemurfenib-resistant) BRAF-mutated melanoma. In some embodiments, the melanoma is relapsed. In some embodiments, the melanoma is refractory.
[00186] In some embodiments, the present invention relates to a method of treating a cancer wherein the method comprises administering to a patient in need thereof a composition comprising Compound 1, or a pharmaceutically acceptable salt thereof, in combination with a TOR inhibitor, wherein the cancer is colorectal cancer. In certain embodiments, the colorectal cancer is locally advanced. In certain embodiments, the colorectal cancer is metastatic. In certain embodiments, the colorectal cancer is a BRAF-mutated colorectal cancer. In certain embodiments, the colorectal cancer is a BRAFv600-mutated colorectal cancer. In certain embodiments, the colorectal cancer is a RAS-mutated colorectal cancer. In certain embodiments, the colorectal cancer is a KRAS-mutated colorectal cancer. In certain embodiments, the colorectal cancer is a NRAS-mutated colorectal cancer. In some embodiments, the colorectal cancer is relapsed. In some embodiments, the colorectal cancer is refractory.
[00187] In some embodiments, the present invention relates to a method of treating a cancer wherein the method comprises administering to a patient in need thereof a composition comprising Compound 1, or a pharmaceutically acceptable salt thereof, in combination with a TOR inhibitor, wherein the cancer is pancreatic cancer. In certain embodiments, the pancreatic cancer is locally advanced. In certain embodiments, the pancreatic cancer is metastatic. In certain embodiments, the pancreatic cancer is a pancreatic ductal adenocarcinoma (PDAC). In certain embodiments, the pancreatic cancer is a RAS-mutated pancreatic cancer. In certain embodiments, the pancreatic cancer is a KRAS-mutated pancreatic cancer. In certain embodiments, the pancreatic cancer is KRAS-mutated pancreatic cancer, including, but not limited to, KRASG12C/D/V, KRASG13C/D,or KRASQ61L/H/R. In some embodiments, the pancreatic cancer is relapsed. In some embodiments, the pancreatic cancer is refractory.
[00188] In some embodiments, the present invention relates to a method of treating a cancer wherein the method comprises administering to a patient in need thereof a composition comprising Compound 1, or a pharmaceutically acceptable salt thereof, in combination with a TOR inhibitor, wherein the cancer is a papillary thyroid cancer. In certain embodiments, the papillary thyroid cancer is locally advanced. In some embodiments, the papillary thyroid cancer is metastatic. In some embodiments, the papillary thyroid cancer is recurring. In some embodiments, the papillary thyroid cancer is BRAF-mutated papillary thyroid cancer. In some embodiments, the papillary thyroid cancer is BRAFv600-mutated papillary thyroid cancer. In some embodiments, the papillary thyroid cancer is relapsed. In some embodiments, the papillary thyroid cancer is refractory. In some embodiments, the papillary thyroid cancer includes undifferentiated or dedifferentiated histology.
[00189] In some embodiments, the present invention relates to a method of treating a cancer wherein the method comprises administering to a patient in need thereof a composition comprising Compound 1, or a pharmaceutically acceptable salt thereof, in combination with a TOR inhibitor, wherein the cancer is lung cancer. In certain embodiments, the lung cancer is non-small cell lung cancer (NSCLC). In certain embodiments, the lung cancer is locally advanced. In certain embodiments, the lung cancer is metastatic. In certain embodiments, the lung cancer is a RAS-mutated lung cancer. In certain embodiments, the lung cancer is KRAS- mutated lung cancer. In certain embodiments, the lung cancer is a KRAS-mutated lung cancer, including, but not limited to, KRASG12C/D/V, KRASG13C/D,or KRASQ61L/H/R. In some embodiments, the lung cancer is relapsed. In some embodiments, the lung cancer is refractory.
[00190] In some embodiments, the present invention relates to a method of treating a cancer wherein the method comprises administering to a patient in need thereof a composition comprising Compound 1, or a pharmaceutically acceptable salt thereof, in combination with a TOR inhibitor, wherein the cancer is a leukemia. In some embodiments, a leukemia is a chronic leukemia. In certain embodiments, a leukemia is chronic myeloid leukemia. In some embodiments, a leukemia is an acute leukemia. In certain embodiments, a leukemia is acute myeloid leukemia (AML). In certain embodiments, a leukemia is acute monocytic leukemia (AMoL, or AML-M5). In certain embodiments, a leukemia is acute lymphoblastic leukemia (ALL). In certain embodiments, a leukemia is acute T cell leukemia. In certain embodiments, a leukemia is myelomonoblastic leukemia. In certain embodiments, a leukemia is human B cell precursor leukemia. In certain embodiments, a leukemia has a Flt3 mutation or rearrangement. In some embodiments, the leukemia is relapsed. In some embodiments, the leukemia is refractory.
[00191] In some embodiments, the present invention relates to a method of treating a cancer wherein the method comprises administering to a patient in need thereof a composition comprising Compound 1, or a pharmaceutically acceptable salt thereof, in combination with a TOR inhibitor, wherein the cancer is a CNS cancer, for instance CNS tumors. In certain embodiments, a CNS tumor is a glioblastoma or glioblastoma multiforme (GBM). In some embodiments, the present invention relates to a method of treating stomach (gastric) and esophageal tumors and cancers.
[00192] In some embodiments, the present invention relates to a method of treating a cancer wherein the method comprises administering to a patient in need thereof a composition comprising Compound 1, or a pharmaceutically acceptable salt thereof, in combination with a TOR inhibitor, wherein the cancer is multiple myeloma (MM). In certain embodiments, the multiple myeloma is locally advanced. In certain embodiments, the multiple myeloma is metastatic. In certain embodiments, the multiple myeloma is a RAS-mutated multiple myeloma. In certain embodiments, the multiple myeloma is KRAS-mutated multiple myeloma. In certain embodiments, the multiple myeloma is a KRAS-mutated multiple myeloma, including, but not limited to, KRASG12C/D/V, KRASG13C/D, or KRASQ61L/H/R. In some embodiments, the multiple myeloma is relapsed. In some embodiments, the multiple myeloma is refractory.
[00193] In some embodiments, the present invention relates to a method of treating a cancer, wherein the method comprises administering to a patient in need thereof a composition comprising Compound 1, or a pharmaceutically acceptable salt thereof, in combination with a TOR inhibitor, wherein the cancer is hepatocellular carcinoma (HCC). In certain embodiments, the HCC is locally advanced. In certain embodiments, the HCC is metastatic. In certain embodiments, the HCC is a RAS-mutated HCC. In certain embodiments, the HCC is KRAS- mutated HCC. In certain embodiments, the HCC is a KRAS-mutated HCC, including, but not limited to, KRASG12C/D/V, KRASG13C/D, or KRASQ61L/H/R. In some embodiments, the hepatocellular carcinoma is relapsed. In some embodiments, the hepatocellular carcinoma is refractory.
[00194] In some embodiments, the present invention relates to a method of treating a cancer wherein the method comprises administering to a patient in need thereof a composition comprising Compound 1, or a pharmaceutically acceptable salt thereof, in combination with a TOR inhibitor, wherein the cancer is selected from breast, colorectal, endometrial, hematological, leukemia (e.g., AML), liver, lung, melanoma, ovarian, pancreatic, prostate, or thyroid.
[00195] In some embodiments, the present invention relates to a method of treating a cancer wherein the method comprises administering to a patient in need thereof a composition comprising Compound 1, or a pharmaceutically acceptable salt thereof, in combination with a TOR inhibitor, wherein the cancer is selected from breast, colorectal, endometrial, liver, lung, melanoma, ovarian, pancreatic, or thyroid.
[00196] In some embodiments, the present invention relates to a method of treating a cancer wherein the method comprises administering to a patient in need thereof a composition comprising Compound 1, or a pharmaceutically acceptable salt thereof, in combination with a TOR inhibitor, wherein the cancer is selected from colorectal, lung, melanoma, or pancreatic.
[00197] In some embodiments, the present invention relates to a method of treating a cancer wherein the method comprises administering to a patient in need thereof a composition comprising Compound 1, or a pharmaceutically acceptable salt thereof, in combination with a TOR inhibitor, wherein the cancer is selected from colorectal, melanoma, or pancreatic.. Combination Dosing
[00198] As described herein, provided methods comprise administration to a patient in need thereof Compound 1, or a pharmaceutically acceptable salt thereof, in combination with a TOR inhibitor. As used herein, the term“in combination” with regard to administration of Compound 1 and a TOR inhibitor means that each of Compound 1 and the TOR inhibitor can be administered to the patient in any order (i.e., simultaneously or sequentially) or together in a single composition, formulation, or unit dosage form.
[00199] It will be appreciated that Compound 1, or a pharmaceutically acceptable salt thereof, and the TOR inhibitor can be administered on the same day or on different days and in any order as according to an appropriate dosing protocol. Dosing of Compound 1
[00200] In some embodiments, the present invention provides a method of treating, stabilizing or lessening the severity or progression of one or more diseases or disorders associated with one or both of ERK1 and ERK2 comprising administering to a patient in need thereof a TOR inhibitor in combination with a particular total daily dose of Compound 1, wherein the total daily dose of Compound 1 is selected from about 5 mg, about 10 mg, about 15 mg, about 20 mg, about 25 mg, about 30 mg, about 35 mg, about 40 mg, about 45 mg, about 50 mg, about 55 mg, about 60 mg, about 65 mg, about 70 mg, about 75 mg, about 80 mg, about 85 mg, about 90 mg, about 95 mg, about 100 mg, about 105 mg, about 110 mg, about 115 mg, about 120 mg, about 125 mg, about 130 mg, about 135 mg, about 140 mg, about 145 mg, about 150 mg, about 155 mg, about 160 mg, about 165 mg, about 170 mg, about 175 mg, about 180 mg, about 185 mg, about 190 mg, about 195 mg, about 200 mg, about 205 mg, about 210 mg, about 215 mg, about 220 mg, about 225 mg, about 230 mg, about 235 mg, about 240 mg, about 245 mg, about 250 mg, about 255 mg, about 260 mg, about 265 mg, about 270 mg, about 275 mg, about 280 mg, about 285 mg, about 290 mg, about 295 mg, about 300 mg, about 305 mg, about 310 mg, about 315 mg, about 320 mg, about 325 mg, about 330 mg, about 335 mg, about 340 mg, about 345 mg, about 350 mg, about 355 mg, about 360 mg, about 365 mg, about 370 mg, about 375 mg, about 380 mg, about 385 mg, about 390 mg, about 395 mg, about 400 mg, about 405 mg, about 410 mg, about 415 mg, about 420 mg, about 425 mg, about 430 mg, about 435 mg, about 440 mg, about 445 mg, about 450 mg, about 455 mg, about 460 mg, about 465 mg, about 470 mg, about 475 mg, about 480 mg, about 485 mg, about 490 mg, about 495 mg, about 500 mg, about 505 mg, about 510 mg, about 515 mg, about 520 mg, about 525 mg, about 530 mg, about 535 mg, about 540 mg, about 545 mg, about 550 mg, about 555 mg, about 560 mg, about 565 mg, about 570 mg, about 575 mg, about 580 mg, about 585 mg, about 590 mg, about 595 mg, about 600 mg, about 605 mg, about 610 mg, about 615 mg, about 620 mg, about 625 mg, about 630 mg, about 635 mg, about 640 mg, about 645 mg, about 650 mg, about 655 mg, about 660 mg, about 665 mg, about 670 mg, about 675 mg, about 680 mg, about 685 mg, about 690 mg, about 695 mg, about 700 mg, about 705 mg, about 710 mg, about 715 mg, about 720 mg, about 725 mg, about 730 mg, about 735 mg, about 740 mg, about 745 mg, about 750 mg, about 760 mg, about 770 mg, about 780 mg, about 790 mg, about 800 mg, about 825 mg, about 850 mg, about 875 mg, about 900 mg, about 925 mg, about 950 mg, about 975 mg, about 1000 mg, about 1025 mg, about 1050 mg, about 1075 mg, about 1100 mg, about 1125 mg, about 1150 mg, about 1175 mg, about 1200 mg, about 1225 mg, about 1250 mg, about 1275 mg, about 1300 mg, about 1325 mg, about 1350 mg, about 1375 mg, about 1400 mg, about 1425 mg, about 1450 mg, about 1475 mg, about 1500 mg, about 1525 mg, about 1550 mg, about 1575 mg, about 1600 mg, about 1625 mg, about 1650 mg, about 1675 mg, about 1700 mg, about 1725 mg, about 1750 mg, about 1775 mg, about 1800 mg, about 1825 mg, about 1850 mg, about 1875 mg, about 1900 mg, about 1925 mg, about 1950 mg, about 1975 mg, about 2000 mg, about 2050 mg, about 2100 mg, about 2150 mg, about 2200 mg, about 2250 mg, about 2300 mg, about 2350 mg, about 2400 mg, about 2450 mg, about 2500 mg, about 2550 mg, about 2600 mg, about 2650 mg, about 2700 mg, about 2750 mg, about 2800 mg, about 2850 mg, about 2900 mg, about 2950 mg, or about 3000 mg.
[00201] In some embodiments, the present invention provides a method of treating, stabilizing or lessening the severity or progression of one or more diseases or disorders associated with one or both of ERK1 and ERK2 comprising administering to a patient in need thereof a TOR inhibitor in combination with a particular total daily dose of Compound 1, wherein the total daily dose of Compound 1 is selected from about 100 mg, about 150 mg, about 200 mg, about 250 mg, about 300 mg, about 350 mg, about 400 mg, about 450 mg, about 500 mg, about 550 mg, about 600 mg, about 650 mg, about 700 mg, about 750 mg, about 800 mg, about 850 mg, about 900 mg, about 950 mg, about 1000 mg, about 1050 mg, about 1100 mg, about 1150 mg, about 1200 mg, about 1250 mg, about 1300 mg, about 1350 mg, about 1400 mg, about 1450 mg, about 1500 mg, about 1550 mg, about 1600 mg, about 1650 mg, about 1700 mg, about 1750 mg, about 1800 mg, about 1850 mg, about 1900 mg, about 1950 mg, about 2000 mg, about 2050 mg, about 2100 mg, about 2150 mg, about 2200 mg, about 2250 mg, about 2300 mg, about 2350 mg, about 2400 mg, about 2450 mg, about 2500 mg, about 2550 mg, about 2600 mg, about 2650 mg, about 2700 mg, about 2750 mg, about 2800 mg, about 2850 mg, about 2900 mg, about 2950 mg, or about 3000 mg.
[00202] In some embodiments, the present invention provides a method of treating, stabilizing or lessening the severity or progression of one or more diseases or disorders associated with one or both of ERK1 and ERK2 comprising administering to a patient in need thereof a TOR inhibitor in combination with a particular total daily dose of Compound 1, wherein the total daily dose of Compound 1 is selected from about 10 mg to about 500 mg, or about 10 mg to about 450 mg, or about 10 mg to about 425 mg, or about 10 mg to about 400 mg, or about 10 mg to about 375 mg, or about 10 mg to about 350 mg, or about 10 mg to about 325 mg, or about10 mg to about 300 mg, or about 10 mg to about 275 mg, or about 10 to about 250 mg, or about 10 to about 225 mg, or about 10 mg to about 200 mg, or about 10 mg to about 190 mg, or about 10 mg to about 180 mg, or about 10 mg to about 170 mg, or about 10 mg to about 160 mg, or about 10 mg to about 150 mg, or about 10 mg to about 140 mg, or about 10 mg to about 130 mg, or about 10 mg to about 120 mg, or about 10 mg to about 110 mg, or about 10 mg to about 100 mg, or about 10 mg to about 90 mg, or about 10 mg to about 80 mg, or about 10 mg to about 70 mg, or about 10 mg to about 60 mg, or about 10 mg to about 50 mg, or about 10 mg to about 40 mg, or about 10 mg to about 30 mg, or about 20 mg to about 40 mg, or about 20 mg to about 60 mg, or about 20 mg to about 80 mg, or about 40 mg to about 200 mg, or about 40 mg to about 160 mg, or about 80 mg to about 320 mg, or about 80 mg to about 160 mg.
[00203] In some embodiments, the present invention provides a method of treating, stabilizing or lessening the severity or progression of one or more diseases or disorders associated with one or both of ERK1 and ERK2 comprising administering to a patient in need thereof a TOR inhibitor in combination with a particular total daily dose of Compound 1, wherein the total daily dose of Compound 1 is selected from about 100 mg to about 3000 mg, or about 500 mg to about 3000 mg, or about 100 mg to about 2500 mg, or about 500 mg to about 2500 mg, or about 100 mg to about 2200 mg, or about 500 mg to about 2200 mg, or about 600 mg to about 2200 mg, or about 700 mg to about 2200 mg, or about 800 to about 2200 mg, or about 800 to about 2100 mg, or about 800 to about 2000 mg. In certain embodiments, the daily dose is about 800 mg to about 2000 mg. [00204] In some embodiments, a total daily dose of Compound 1 is administered once daily (QD), wherein the dose is selected from about 5 mg, about 10 mg, about 20 mg, about 40 mg, about 80 mg, about 120 mg, about 180 mg, about 330 mg, about 480 mg, or about 640 mg.
[00205] In some embodiments, a total daily dose of Compound 1 is administered once daily (QD), wherein the dose is selected from about 20 mg, about 40 mg, about 80 mg, or about 160 mg. Dosing of a TOR inhibitor
[00206] A dose of a TOR inhibitor to be administered to a patient is rather widely variable and can be patient to the judgment of a health-care practitioner. In general, TOR inhibitors can be administered one to four times a day in a dose of about 0.005 mg/kg of a patient’s body weight to about 10 mg/kg of a patient’s body weight in a patient, but the above dosage may be properly varied depending on the age, body weight and medical condition of the patient and the type of administration. In some embodiments, the dose is about 0.01 mg/kg of a patient’s body weight to about 5 mg/kg of a patient’s body weight, about 0.05 mg/kg of a patient’s body weight to about 1 mg/kg of a patient’s body weight, about 0.1 mg/kg of a patient’s body weight to about 0.75 mg/kg of a patient’s body weight or about 0.25 mg/kg of a patient’s body weight to about 0.5 mg/kg of a patient’s body weight. In some embodiments, one dose is given per day In some embodiments, two doses are given per day. In any given case, the amount of TOR inhibitor administered will depend on such factors as the solubility of the active component, the formulation used and the route of administration.
[00207] In some embodiments, the present invention provides methods for treating, stabilizing or lessening the severity or progression of one or more diseases or disorders associated with one or both of ERK1 and ERK2, comprising administering to a patient in need thereof a composition comprising Compound 1, or a pharmaceutically acceptable salt thereof, and a TOR inhibitor, wherein the TOR inhibitor is administered in an amount of about 0.1 mg/day to about 1200 mg/day, about 1 mg/day to about 100 mg/day, about 10 mg/day to about 1200 mg/day, about 10 mg/day to about 100 mg/day, about 100 mg/day to about 1200 mg/day, about 400 mg/day to about 1200 mg/day, about 600 mg/day to about 1200 mg/day, about 400 mg/day to about 800 mg/day or about 600 mg/day to about 800 mg/day. [00208] In some embodiments, methods disclosed herein comprise the administration of about 0.1 mg/day, about 0.5 mg/day, about 1 mg/day, about 10 mg/day, about 15 mg/day, about 20 mg/day, about 30 mg/day, about 40 mg/day, about 45 mg/day, about 50 mg/day, about 60 mg/day, about 75 mg/day, about 100 mg/day, about 125 mg/day, about 150 mg/day, about 200 mg/day, about 250 mg/day, about 300 mg/day, about 400 mg/day, about 600 mg/day or about 800 mg/day of a TOR inhibitor to a patient in need thereof.
[00209] In some embodiments, methods disclosed herein comprise the administration of about 10 mg/day, about 15 mg/day, about 20 mg/day, about 30 mg/day or about 45 mg/day.
[00210] In some embodiments, methods disclosed herein comprise the administration of about 10 mg/day, about 15 mg/day, about 16 mg/day, about 20 mg/day, about 30 mg/day. Unit Dosage Forms of Compound 1
[00211] Compound 1, or a pharmaceutically acceptable salt thereof, is preferably formulated in unit dosage form for ease of administration and uniformity of dosage. It will be understood, however, that the total daily usage of Compound 1, or a pharmaceutically acceptable salt thereof, and compositions thereof, will be decided by the attending physician within the scope of sound medical judgment. The specific effective dose level for any particular patient or organism will depend upon a variety of factors including the disorder being treated and the severity of the disorder; the specific composition employed; the age, body weight, general health, sex and diet of the patient; the time of administration, route of administration, and rate of excretion of Compound 1; the duration of the treatment; drugs used in combination or coincidental with Compound 1, and like factors well known in the medical arts. A person of ordinary skill will appreciate that the unit dosage forms described herein refer to an amount of Compound 1, i.e. the free base form of the active pharmaceutical ingredient, which may be provided as the free base or as a pharmaceutically acceptable salt thereof.
[00212] In some embodiment, the present invention provides methods for treating, stabilizing or lessening the severity or progression of one or more diseases or disorders associated with one or both of ERK1 and ERK2, comprising administering to a patient in need thereof a composition comprising Compound 1, or a pharmaceutically acceptable salt thereof, and a TOR inhibitor, wherein Compound 1 is administered in unit dosage formulations that comprise between about about 5 mg to about 1000 mg of Compound 1. In certain embodiments, a unit dosage formulation of the present invention provides about 1 mg, 5 mg, about 10 mg, about 15 mg, about 20 mg, about 25 mg, about 30 mg, about 35 mg, about 40 mg, about 45 mg, about 50 mg, about 55 mg, about 60 mg, about 65 mg, about 70 mg, about 75 mg, about 80 mg, about 85 mg, about 90 mg, about 95 mg, about 100 mg, about 105 mg, about 110 mg, about 115 mg, about 120 mg, about 125 mg, about 130 mg, about 135 mg, about 140 mg, about 145 mg, about 150 mg, about 155 mg, about 160 mg, about 165 mg, about 170 mg, about 175 mg, about 180 mg, about 185 mg, about 190 mg, about 195 mg, about 200 mg, about 225 mg, about 250 mg, about 275 mg, about 300 mg, about 325 mg, about 350 mg, about 375 mg, about 400 mg, about 425 mg, about 450 mg, about 475 mg, about 500 mg, about 550 mg, about 600 mg, about 650 mg, about 700 mg, about 750 mg, about 800 mg, about 850 mg, about 900 mg, about 950 mg, or about 1000 mg of Compound 1.
[00213] In some embodiments, the present invention provides methods for treating, stabilizing or lessening the severity or progression of one or more diseases or disorders associated with one or both of ERK1 and ERK2, comprising administering to a patient in need thereof a composition comprising Compound 1, or a pharmaceutically acceptable salt thereof, and a TOR inhibitor, wherein Compound 1 is administered in unit dosage formulations that comprise about 5 mg, 30 mg, or 150 mg of Compound 1. In certain embodiments, a capsule formulation of the present invention provides about 25 mg, about 50 mg, about 75 mg, about 100 mg, about 125 mg, or about 150 mg of Compound 1.
[00214] In certain embodiments, Compound 1, or a pharmaceutically acceptable salt thereof, is administered at dosage levels of about 0.01 mg/kg to about 50 mg/kg and preferably from about 1 mg/kg to about 25 mg/kg, of subject body weight per day, one or more times a day, to obtain the desired therapeutic effect. Unit Dosage Forms of a TOR Inhibitor
[00215] In some embodiment, the present invention provides methods for treating, stabilizing or lessening the severity or progression of one or more diseases or disorders associated with one or both of ERK1 and ERK2, comprising administering to a patient in need thereof a composition comprising Compound 1, or a pharmaceutically acceptable salt thereof, and a TOR inhibitor, wherein the TOR inhibitor is administered in unit dosage formulations that comprise between about 0.1 mg and about 2000 mg, about 1 mg and 200 mg, about 35 mg and about 1400 mg, about 125 mg and about 1000 mg, about 250 mg and about 1000 mg, or about 500 mg and about 1000 mg of a TOR inhibitor.
[00216] In some embodiments, provided herein are unit dosage formulations comprising about 0.1 mg, 0.25 mg, 0.5 mg, 1 mg, 5 mg, 10 mg, 15 mg, 20 mg, 30 mg, 45 mg, 50 mg, 60 mg, 75 mg, 100 mg, 125 mg, 150 mg, 200 mg, 250 mg, 300 mg, 400 mg, 600 mg or 800 mg of a TOR inhibitor.
[00217] In some embodiments, provided herein are unit dosage formulations that comprise 0.1 mg, 0.25 mg, 0.5 mg, 1 mg, 2.5 mg, 5 mg, 10 mg, 15 mg, 20 mg, 30 mg, 35 mg, 50 mg, 70 mg, 100 mg, 125 mg, 140 mg, 175 mg, 200 mg, 250 mg, 280 mg, 350 mg, 500 mg, 560 mg, 700 mg, 750 mg, 1000 mg or 1400 mg of a TOR inhibitor. In a particular embodiment, provided herein are unit dosage formulations that comprise about 5 mg, about 15 mg, about 20 mg, about 30 mg, about 45 mg, and about 50 mg of a TOR inhibitor.
[00218] In some embodiments, provided herein are unit dosage formulations that comprise about 2 mg, about 5 mg, about 7.5 mg, about 10 mg and about 15 mg of a TOR inhibitor. Administration of Compound 1
[00219] Compound 1, or a pharmaceutically acceptable salt thereof, and compositions thereof according to methods of the present invention, are administered using any amount and any route of administration effective for treating or lessening the severity of a disorder provided above. The exact amount required will vary from subject to subject, depending on the species, age, and general condition of the subject, the severity of the infection, the particular agent, its mode of administration, and the like.
[00220] In some embodiments, provided methods comprise administering a pharmaceutically acceptable composition comprising Compound 1, or a pharmaceutically acceptable salt thereof, one, two, three, or four times a day.
[00221] In some embodiments, a pharmaceutically acceptable composition comprising Compound 1, or a pharmaceutically acceptable salt thereof, is administered once daily (“QD”). [00222] In some embodiments, a pharmaceutically acceptable composition comprising Compound 1, or a pharmaceutically acceptable salt thereof, is administered twice daily. In some embodiments, twice daily administration refers to a compound or composition that is administered“BID”, or two equivalent doses administered at two different times in one day.
[00223] In some embodiments, a pharmaceutically acceptable composition comprising Compound 1, or a pharmaceutically acceptable salt thereof, is administered three times a day. In some embodiments, a pharmaceutically acceptable composition comprising Compound 1, or a pharmaceutically acceptable salt thereof, is administered“TID”, or three equivalent doses administered at three different times in one day.
[00224] In some embodiments, a pharmaceutically acceptable composition comprising Compound 1, or a pharmaceutically acceptable salt thereof, is administered four times a day. In some embodiments, a pharmaceutically acceptable composition comprising Compound 1, or a pharmaceutically acceptable salt thereof, is administered“QID”, or four equivalent doses administered at four different times in one day.
[00225] In some embodiments, Compound 1 is administered to a patient under fasted conditions and the total daily dose is any of those contemplated above and herein.
[00226] In some embodiments, Compound 1 is administered to a patient under fed conditions and the total daily dose is any of those contemplated above and herein.
[00227] In some embodiments, Compound 1 is administered orally.
[00228] Pharmaceutically acceptable compositions of this invention can be administered to humans and other animals orally, rectally, parenterally, intracisternally, intravaginally, intraperitoneally, topically (as by powders, ointments, or drops), bucally, as an oral or nasal spray, or the like, depending on the severity of the disease or disorder being treated. Administration of a TOR Inhibitor
[00229] In some embodiments, provided methods comprise administering a pharmaceutically acceptable composition comprising a TOR inhibitor one, two, three, or four times a day.
[00230] In some embodiments, a pharmaceutically acceptable composition comprising a TOR inhibitor is administered once daily (“QD”). [00231] In some embodiments, a pharmaceutically acceptable composition comprising a TOR inhibitor is administered twice daily. In some embodiments, twice daily administration refers to a compound or composition that is administered“BID”, or two equivalent doses administered at two different times in one day.
[00232] In some embodiments, a pharmaceutically acceptable composition comprising a TOR inhibitor is administered three times a day. In some embodiments, a pharmaceutically acceptable composition comprising a TOR inhibitor is administered“TID”, or three equivalent doses administered at three different times in one day.
[00233] In some embodiments, a pharmaceutically acceptable composition comprising a TOR inhibitor is administered four times a day. In some embodiments, a pharmaceutically acceptable composition comprising a TOR inhibitor is administered“QID”, or four equivalent doses administered at four different times in one day.
[00234] In some embodiments, a TOR inhibitor is administered to a patient under fasted conditions and the total daily dose is any of those contemplated above and herein.
[00235] In some embodiments, a TOR inhibitor is administered to a patient under fed conditions and the total daily dose is any of those contemplated above and herein.
[00236] In some embodiments, a TOR inhibitor is administered orally for reasons of convenience. In some embodiments, when administered orally, a TOR inhibitor is administered with a meal and water. In another embodiment, the TOR inhibitor is dispersed in water or juice (e.g., apple juice or orange juice) and administered orally as a suspension. In some embodiments, when administered orally, a TOR inhibitor is administered in a fasted state.
[00237] A TOR inhibitor can also be administered intradermally, intramuscularly, intraperitoneally, percutaneously, intravenously, subcutaneously, intranasally, epidurally, sublingually, intracerebrally, intravaginally, transdermally, rectally, mucosally, by inhalation, or topically to the ears, nose, eyes, or skin. The mode of administration is left to the discretion of the health-care practitioner, and can depend in-part upon the site of the medical condition. Pharmaceutically Acceptable Compositions of Compound 1
[00238] In some embodiments, the present invention provides a pharmaceutically acceptable composition comprising Compound 1, or a pharmaceutically acceptable salt thereof. In some embodiments, the present invention provides a pharmaceutically acceptable composition of a TOR inhibitor. In some embodiments, a composition comprising Compound 1, or a pharmaceutically acceptable salt thereof, is separate from a composition comprising a TOR inhibitor. In some embodiments, Compound 1, or a pharmaceutically acceptable salt thereof, and a TOR inhibitor are present in the same composition.
[00239] Exemplary such pharmaceutically acceptable compositions are described further below and herein.
[00240] Liquid dosage forms for oral administration include, but are not limited to, pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups and elixirs. In addition to Compound 1, or a pharmaceutically acceptable salt thereof, and/or a TOR inhibitor, the liquid dosage forms may contain inert diluents commonly used in the art such as, for example, water or other solvents, solubilizing agents and emulsifiers such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, dimethylformamide, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor, and sesame oils), glycerol, tetrahydrofurfuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof. Besides inert diluents, the oral compositions can also include adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, and perfuming agents.
[00241] Injectable preparations, for example, sterile injectable aqueous or oleaginous suspensions may be formulated according to the known art using suitable dispersing or wetting agents and suspending agents. The sterile injectable preparation may also be a sterile injectable solution, suspension or emulsion in a nontoxic parenterally acceptable diluent or solvent, for example, as a solution in 1,3-butanediol. Among the acceptable vehicles and solvents that may be employed are water, Ringer's solution, U.S.P. and isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose any bland fixed oil can be employed including synthetic mono- or diglycerides. In addition, fatty acids such as oleic acid are used in the preparation of injectables.
[00242] Injectable formulations can be sterilized, for example, by filtration through a bacterial-retaining filter, or by incorporating sterilizing agents in the form of sterile solid compositions which can be dissolved or dispersed in sterile water or other sterile injectable medium prior to use.
[00243] In order to prolong the effect of Compound 1, and/or a TOR inhibitor, it is often desirable to slow absorption from subcutaneous or intramuscular injection. This may be accomplished by the use of a liquid suspension of crystalline or amorphous material with poor water solubility. The rate of absorption then depends upon its rate of dissolution that, in turn, may depend upon crystal size and crystalline form. Alternatively, delayed absorption of parenterally administered Compound 1, or a pharmaceutically acceptable salt thereof, and/or a TOR inhibitor, is accomplished by dissolving or suspending the compound in an oil vehicle. Injectable depot forms are made by forming microencapsule matrices of Compound 1, or a pharmaceutically acceptable salt thereof, and/or a TOR inhibitor, in biodegradable polymers such as polylactide-polyglycolide. Depending upon the ratio of compound to polymer and the nature of the particular polymer employed, the rate of compound release can be controlled. Examples of other biodegradable polymers include poly(orthoesters) and poly(anhydrides). Depot injectable formulations are also prepared by entrapping the compound in liposomes or microemulsions that are compatible with body tissues.
[00244] Compositions for rectal or vaginal administration are preferably suppositories which can be prepared by mixing the compounds of this invention with suitable non-irritating excipients or carriers such as cocoa butter, polyethylene glycol or a suppository wax which are solid at ambient temperature but liquid at body temperature and therefore melt in the rectum or vaginal cavity and release the active compound.
[00245] Solid dosage forms for oral administration include capsules, tablets, pills, powders, and granules. In such solid dosage forms, the active compound is mixed with at least one inert, pharmaceutically acceptable excipient or carrier such as sodium citrate or dicalcium phosphate and/or a) fillers or extenders such as starches, lactose, sucrose, glucose, mannitol, and silicic acid, b) binders such as, for example, carboxymethylcellulose, alginates, gelatin, polyvinylpyrrolidinone, sucrose, and acacia, c) humectants such as glycerol, d) disintegrating agents such as agar--agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and sodium carbonate, e) solution retarding agents such as paraffin, f) absorption accelerators such as quaternary ammonium compounds, g) wetting agents such as, for example, cetyl alcohol and glycerol monostearate, h) absorbents such as kaolin and bentonite clay, and i) lubricants such as talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate, and mixtures thereof. In the case of capsules, tablets and pills, the dosage form may also comprise buffering agents.
[00246] Solid compositions of a similar type may also be employed as fillers in soft and hard- filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polyethylene glycols and the like. The solid dosage forms of tablets, dragees, capsules, pills, and granules can be prepared with coatings and shells such as enteric coatings and other coatings well known in the pharmaceutical formulating art. They may optionally contain opacifying agents and can also be of a composition that they release the active ingredient(s) only, or preferentially, in a certain part of the intestinal tract, optionally, in a delayed manner. Examples of embedding compositions that can be used include polymeric substances and waxes. Solid compositions of a similar type may also be employed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polethylene glycols and the like.
[00247] Compound 1, or a pharmaceutically acceptable salt thereof, and/or a TOR inhibitor, can also be in micro-encapsulated form with one or more excipients as noted above. The solid dosage forms of tablets, dragees, capsules, pills, and granules can be prepared with coatings and shells such as enteric coatings, release controlling coatings and other coatings well known in the pharmaceutical formulating art. In such solid dosage forms Compound 1, or a pharmaceutically acceptable salt thereof, and/or a TOR inhibitor, may be admixed with at least one inert diluent such as sucrose, lactose or starch. Such dosage forms may also comprise, as is normal practice, additional substances other than inert diluents, e.g., tableting lubricants and other tableting aids such a magnesium stearate and microcrystalline cellulose. In the case of capsules, tablets and pills, the dosage forms may also comprise buffering agents. They may optionally contain opacifying agents and can also be of a composition that they release the active ingredient(s) only, or preferentially, in a certain part of the intestinal tract, optionally, in a delayed manner. Examples of embedding compositions that can be used include polymeric substances and waxes.
[00248] Dosage forms for topical or transdermal administration of Compound 1, or a pharmaceutically acceptable salt thereof, and/or a TOR inhibitor, include ointments, pastes, creams, lotions, gels, powders, solutions, sprays, inhalants or patches. The active component is admixed under sterile conditions with a pharmaceutically acceptable carrier and any needed preservatives or buffers as may be required. Ophthalmic formulation, ear drops, and eye drops are also contemplated as being within the scope of this invention. Additionally, the present invention contemplates the use of transdermal patches, which have the added advantage of providing controlled delivery of a compound to the body. Such dosage forms can be made by dissolving or dispensing the compound in the proper medium. Absorption enhancers can also be used to increase the flux of the compound across the skin. The rate can be controlled by either providing a rate controlling membrane or by dispersing the compound in a polymer matrix or gel.
[00249] According to one embodiment, the invention relates to a method of inhibiting protein kinase activity in a biological sample comprising the step of contacting said biological sample with Compound 1, or a pharmaceutically acceptable salt thereof, and/or a TOR inhibitor, or a composition comprising said compound.
[00250] According to another embodiment, the invention relates to a method of inhibiting one or both of ERK 1 and ERK2 kinase, or a mutant thereof, activity in a biological sample comprising the step of contacting said biological sample with Compound 1, or a pharmaceutically acceptable salt thereof, in combination with a TOR inhibitor, or a composition comprising any of the foregoing. In certain embodiments, the invention relates to a method of irreversibly inhibiting one or both of ERK1 and ERK2 kinase, or a mutant thereof, activity in a biological sample comprising the step of contacting said biological sample with Compound 1, or a pharmaceutically acceptable salt thereof, in combination with a TOR inhibitor, or a composition comprising any of the foregoing.
[00251] The term“biological sample”, as used herein, includes, without limitation, cell cultures or extracts thereof; biopsied material obtained from a mammal or extracts thereof; and blood, saliva, urine, feces, semen, tears, or other body fluids or extracts thereof.
[00252] Inhibition of one or both of ERK1 and ERK2, or a mutant thereof, activity in a biological sample is useful for a variety of purposes that are known to one of skill in the art. Examples of such purposes include, but are not limited to, blood transfusion, organ- transplantation, biological specimen storage, and biological assays. [00253] Another embodiment of the present invention relates to a method of inhibiting protein kinase activity in a patient comprising the step of administering to said patient Compound 1, or a pharmaceutically acceptable salt thereof, in combination with a TOR inhibitor, or a composition comprising any of the foregoing.
[00254] According to certain embodiments, the invention relates to a method of irreversibly inhibiting one or both of ERK1 and ERK2 kinase, or a mutant thereof, activity in a patient comprising the step of administering to said patient Compound 1, or a pharmaceutically acceptable salt thereof, in combination with a TOR inhibitor, or a composition comprising any of the foregoing.
[00255] In certain embodiments, the activity is inhibited irreversibly by covalently modifying Cys 183 of ERK1. In certain embodiments, the activity is inhibited irreversibly by covalently modifying Cys 166 of ERK2. In certain embodiments, the activity is inhibited irreversibly by covalently modifying Cys 183 of ERK1 and Cys 166 of ERK2. Pharmaceutically Acceptable Compositions of TOR Inhibitors
[00256] Provided herein for use in combination with Compound 1, or a pharmaceutically acceptable salt thereof, are compositions comprising an effective amount of a TOR inhibitor and compositions comprising an effective amount of a TOR inhibitor and a pharmaceutically acceptable carrier or vehicle. In some embodiments, the pharmaceutical compositions described herein are suitable for oral, parenteral, mucosal, transdermal or topical administration.
[00257] In some embodiments, TOR inhibitors can be administered to a patient orally or parenterally in the conventional form of preparations, such as capsules, microcapsules, tablets, granules, powder, troches, pills, suppositories, injections, suspensions and syrups. Suitable formulations can be prepared by methods commonly employed using conventional, organic or inorganic additives, such as an excipient (e.g., sucrose, starch, mannitol, sorbitol, lactose, glucose, cellulose, talc, calcium phosphate or calcium carbonate), a binder (e.g., cellulose, methylcellulose, hydroxymethylcellulose, polypropylpyrrolidone, polyvinylpyrrolidone, gelatin, gum arabic, polyethyleneglycol, sucrose or starch), a disintegrator (e.g., starch, carboxymethylcellulose, hydroxypropylstarch, low substituted hydroxypropylcellulose, sodium bicarbonate, calcium phosphate or calcium citrate), a lubricant (e.g., magnesium stearate, light anhydrous silicic acid, talc or sodium lauryl sulfate), a flavoring agent (e.g., citric acid, menthol, glycine or orange powder), a preservative (e.g, sodium benzoate, sodium bisulfite, methylparaben or propylparaben), a stabilizer (e.g., citric acid, sodium citrate or acetic acid), a suspending agent (e.g., methylcellulose, polyvinyl pyrroliclone or aluminum stearate), a dispersing agent (e.g., hydroxypropylmethylcellulose), a diluent (e.g., water), and base wax (e.g., cocoa butter, white petrolatum or polyethylene glycol). An effective amount of a TOR inhibitor in a pharmaceutical composition may be at a level that will exercise the desired effect; for example, about 0.005 mg/kg of a patient’s body weight to about 10 mg/kg of a patient’s body weight in unit dosage for both oral and parenteral administration.
[00258] In some embodiments, provided herein are capsules containing a TOR inhibitor without an additional carrier, excipient or vehicle.
[00259] In some embodiments, provided herein are compositions comprising an effective amount of a TOR inhibitor and a pharmaceutically acceptable carrier or vehicle, wherein a pharmaceutically acceptable carrier or vehicle can comprise an excipient, diluent, or a mixture thereof. In some embodiments, the composition is a pharmaceutical composition.
[00260] The compositions can be in the form of tablets, chewable tablets, capsules, solutions, parenteral solutions, troches, suppositories and suspensions and the like. Compositions can be formulated to contain a daily dose, or a convenient fraction of a daily dose, in a dosage unit, which may be a single tablet or capsule or convenient volume of a liquid. In some embodiments, the solutions are prepared from water-soluble salts, such as the hydrochloride salt. In general, all of the compositions are prepared according to known methods in pharmaceutical chemistry. Capsules can be prepared by mixing a TOR inhibitor with a suitable carrier or diluent and filling the proper amount of the mixture in capsules. The usual carriers and diluents include, but are not limited to, inert powdered substances such as starch of many different kinds, powdered cellulose, especially crystalline and microcrystalline cellulose, sugars such as fructose, mannitol and sucrose, grain flours and similar edible powders.
[00261] Tablets can be prepared by direct compression, by wet granulation, or by dry granulation. Their formulations usually incorporate diluents, binders, lubricants and disintegrators as well as the compound. Typical diluents include, for example, various types of starch, lactose, mannitol, kaolin, calcium phosphate or sulfate, inorganic salts such as sodium chloride and powdered sugar. Powdered cellulose derivatives are also useful. In some embodiments, the pharmaceutical composition is lactose-free. Typical tablet binders are substances such as starch, gelatin and sugars such as lactose, fructose, glucose and the like. Natural and synthetic gums are also convenient, including acacia, alginates, methylcellulose, polyvinylpyrrolidine and the like. Polyethylene glycol, ethylcellulose and waxes can also serve as binders.
[00262] A lubricant might be necessary in a tablet formulation to prevent the tablet and punches from sticking in the die. The lubricant can be chosen from such slippery solids as talc, magnesium and calcium stearate, stearic acid and hydrogenated vegetable oils. Tablet disintegrators are substances that swell when wetted to break up the tablet and release the compound. They include starches, clays, celluloses, algins and gums. More particularly, corn and potato starches, methylcellulose, agar, bentonite, wood cellulose, powdered natural sponge, cation-exchange resins, alginic acid, guar gum, citrus pulp and carboxymethyl cellulose, for example, can be used as well as sodium lauryl sulfate. Tablets can be coated with sugar as a flavor and sealant, or with film-forming protecting agents to modify the dissolution properties of the tablet. The compositions can also be formulated as chewable tablets, for example, by using substances such as mannitol in the formulation.
[00263] When it is desired to administer a TOR inhibitor as a suppository, typical bases can be used. Cocoa butter is a traditional suppository base, which can be modified by addition of waxes to raise its melting point slightly. Water-miscible suppository bases comprising, particularly, polyethylene glycols of various molecular weights are in wide use.
[00264] The effect of the TOR inhibitor can be delayed or prolonged by proper formulation. For example, a slowly soluble pellet of the TOR inhibitor can be prepared and incorporated in a tablet or capsule, or as a slow-release implantable device. The technique also includes making pellets of several different dissolution rates and filling capsules with a mixture of the pellets. Tablets or capsules can be coated with a film that resists dissolution for a predictable period of time. Even the parenteral preparations can be made long-acting, by dissolving or suspending the TOR inhibitor in oily or emulsified vehicles that allow it to disperse slowly in the serum. Additional formulations can be found in USSN 14/288,521, USSN 14/254,023, and USSN 62/003,173, the entirety of each of which is hereby incorporated by reference. [00265] All features of each of the aspects of the invention apply to all other aspects mutatis mutandis. Each of the references referred to herein, including but not limited to patents, patent applications and journal articles, is incorporated by reference herein as though fully set forth in its entirety.
[00266] In order that the invention described herein may be more fully understood, the following examples are set forth. It should be understood that these examples are for illustrative purposes only and are not to be construed as limiting this invention in any manner. EXEMPLIFICATION
Example 1
General Preparation of Compound 1
[00267] As depicted in the Examples below, in certain exemplary embodiments, Compound 1 is prepared according to the following general procedure.
[00268] Proton Nuclear Magnetic Resonance (1H NMR) spectra were obtained on a Bruker AVANCE-300 MHz NMR spectrometer.Deuterated DMSO was used as solvent.
Figure imgf000072_0001
[00269] The title compound was prepared according to the steps and intermediates described below and in the‘230 publication, the entirety of which is incorporated herein by reference. Step 1: N-(2-(2-Chloro-5-(trifluoromethyl)pyrimidin-4-ylamino)5- methylphenyl)acrylamide (Intermediate 1)
Figure imgf000073_0001
[00270] To a stirred solution of N-(2-amino-5-methylphenyl)acrylamide (22.2 mmol) in dimethyl acetamide (25 mL) was added potassium carbonate (46.0 mmol) at rt, and the mixture was stirred for 15 minutes. To this reaction mixture, 2,4-dichloro-5-trifluoromethylpyrimidine (22.2 mmol) was added, and the stirring continued at 60 oC for 1 h. Upon completion, the reaction mixture was diluted with water (2x50 mL) and extracted with EtOAc (2x100 mL). The organic layer was dried over sodium sulfate and concentrated to get the crude product. This crude was purified by silica gel column chromatography and subsequently purified by prep- HPLC to get desired Intermediate 1. Step 2: Acid catalyzed coupling method
[00271] To a solution of Intermediate 1 (2.923 mmol) in 0.04 M PTSA solution in 1,4- dioxane (20 mL) was added 2-methoxy-5-methylpyridin-4-amine (3.5076 mmol), and the mixture was stirred at 95 oC for 16 h. Upon completion, the reaction mixture was directly absorbed on silica gel and purified by column chromatography. The resulting product was stirred in a mixture of DCM: EtOAc: diethyl ether (10 mL:10 mL:30 mL) for 10 min, then filtered and dried under vacuum to obtain the desired compound.
[00272] MS m/z 459.2 (ES+, M+H). 1HNMR (DMSO-d6) δ 2.10 (s, 3H), 2.32 (s, 3H), 3.75 (s, 3H), 5.78 (dd, 1H, J = 2.0, 10.0 Hz), 6.28 (dd, 1H, J = 2.0, 16.8 Hz), 6.45 (dd, 1H, J = 10.6, 16.8 Hz), 7.09 (br t, 3 H, J = 8.0 Hz), 7.50 (d, 1H, J = 8.4 Hz), 7.79 (s, 1H), 8.36 (s, 2H), 8.72 (s, 1H), 10.25 (s, 1H).
Alternative Step 2: Pd-catalyzed coupling method:
[00273] Alternatively, Step 2 can be carried out by adding Intermediate 1 to a suitable coupling partner in the presence of Na2CO3, a degassed solvent (e.g., tert-amyl alcohol), a suitable palladium catalyst (e.g., tris-dibenzylamino dipalladium) and a suitable phosphine ligand (e.g., Dave Phos) under conditions suitable to effect coupling. Example 2
In Vitro Combination Therapy Assays
[00274] This Example describes experiments relating to the effect of the combination of Compound 1 (free base) with Compound 2 on the Panc1 (a.k.a. PANC-1) pancreatic cancer cell line, the Mia PaCa pancreatic cancer cell line, the HS294T melanoma cell line, the HCT-116 colorectal cancer cell line, the NCI-H460 lung cancer cell line, the NCI-H522 lung cancer cell line, or the NCI-H1755 lung cancer cell line.
[00275] For dose-response viability assays, cells were plated at a density of 3000 cells/well in 90 µL of growth media on 96 well clear bottom black-well plates (Corning Cat# 3904) and incubated overnight under standard cell culture growth conditions at 37 0C 5% CO2. The outer most rows and columns of wells were filled with culture media, without cells, to avoid evaporation effects on subsequent readouts.
[00276] The following day, one plate for each cell line was used for“Day 0” cell growth control readout, and cell viability was measured with CellTiter Glo (Promega) reagent according to manufacturer specifications. Day 0 control plates were equilibrated to room temperature for 30 minutes in a tissue culture hood, followed by addition of 90 µL of CellTiter Glo reagent, a volume equal to cell media volume, to each well. The plates were covered with foil to protect from light and placed on a shaker at low speed for 5 minutes, followed by 10 minutes incubation without shaking. CellTiter Glo reagent signal was read on a spectramax L luminescence detector, and data were processed using Excel and Prism software.
[00277] At the same time as the“Day 0” plate was equilibrating to room temperature, cells in the remaining plates were treated with Compound 1 alone, Compound 2, a combination thereof, or DMSO vehicle control, as specified for each cell line. For cell treatment compounds were diluted from starting concentration 10 mM stock in DMSO to 10 µM starting in-well concentration (3 dilution steps first serially in DMSO, then in cell growth media), such that resulting DMSO concentration in the growth media in wells containing cells was 0.1%. The cells were treated with either 9 point 3-fold dilutions of Compound 1 alone, or 9 point 3-fold dilutions of Compound 2 alone, or a combination of Compound 1 and Compound 2, or DMSO vehicle control. Thus, the final concentrations in treatment wells for each compound was: (in nM) 10000.00, 3333.33, 1111.11, 370.37, 123.46, 41.15, 13.72, 4.57, 1.52, 0. Each treatment was contained in 10 µL treatment media added to the respective wells. Each concentration was tested in triplicate.
[00278] Seventy two hours later“Day 3” cell viability was measured by adding 100 µL CellTiter Glo reagent to each of the treatment wells, equal to the volume of media and compound. Cell viability was assessed as described for“Day 0” control. Summary of Results:
Effects of Compound 1 and Compound 2 Used Alone or in Combination with Each Other on Viability of Panc1 Pancreatic Cancer Cell Line (see Figure 1).
[00279] Figure 1. Panc1 (a.k.a., PANC-1) Pancreatic Cancer Cell Line: Panc 1 cells were treated with a 9-point dose dilution of Compound 1 or Compound 2 or both, ranging from 1.52 nM to 10000.00 nM and DMSO vehicle control. Fig. 1A: dose response to Compound 2 (trend line with triangles); Fig. 1B: dose response to Compound 1 (trend line with circles); Fig. 1C: combination treatment with Compound 1 and Compound 2 (trend line with circles).
[00280] Summary: Compound 2 induces cytostatic (growth-inhibitory) effect but not cell death, as noted by the dose-response curve remaining above the X-axis. Compound 1 induces cell-death (cytotoxic) effect (noted by the curve crossing the X-axis) at 1111.11 nM, and the combination of Compound 1 and Compound 2 induces a cell death effect of the same magnitude as Compound 1 (since the curves plateau at the same level below x-axis between -30.1% and - 34.5% of starting cell growth control) but at a lower dose (370.37 nM of each agent). Effects of Compound 1 and Compound 2 Used Alone or in Combination with Each Other on Viability of Mia PaCa Pancreatic Cancer Cell Line (Figure 2).
[00281] Figure 2. Mia PaCa Pancreatic Cancer Cell Line: Mia PaCa cells were treated with a 9-point dose dilution of Compound 1, or Compound 2, or both, ranging from 1.52 nM to 10000.00 nM, and DMSO vehicle control. Fig. 2A: dose response to Compound 2; Fig. 2B: dose response to Compound 1 (trend line with circles); Fig. 2C: combination treatment with Compound 1 and Compound 2 (trend line with circles).
[00282] Summary: Compound 2 induces cytostatic (growth-inhibitory) effect but not cell death, as noted by the curve remaining above the X-axis. Compound 1 induces cell death (noted by the curve crossing the X-axis) at 247 nM, and the combination of Compound 1 and Compound 2 induces a cell death effect of increased magnitude (since the curve of the Compound 1 and Compound 2 combination response plateaus at the -84.6% of starting growth control compared to -49.8% induced by Compound 1 alone) at a dose of 123.46 nM of each agent combined. Effects of Compound 1 and Compound 2 Used Alone or in Combination with Each Other on Viability of HS294T Melanoma Cell Line (Figure 3).
[00283] Figure 3. HS294T were treated with a 9-point dose dilution of Compound 1, Compound 2, or both, ranging from 1.52 nM to 10000.00 nM and DMSO vehicle control. Fig. 3A: dose response to Compound 2 (trend line with triangles); Fig. 3B: dose response to Compound 1 (trend line with circles); Fig. 3C: combination treatment with Compound 1 and Compound 2 (trend line with circles).
[00284] Summary: Either Compound 1 or Compound 2 alone induce cell death at dose of 370.37 nM of either inhibitor. The magnitude of cell death response is higher for Compound 1 at -88% of starting growth control. The combination of Compound 1 and Compound 2 induces cell death of magnitude similar to that of Compound 1 alone, but at lower dose (123.46 nM of each inhibitor). Effects of Compound 1 and Compound 2 Used Alone or in Combination with Each Other on Viability of HCT-116 Colorectal Cell Line (Figure 4).
[00285] Figure 4. HCT-116 cells were treated with a 9-point dose dilution of Compound 1 (Fig. 4A) or Compound 2 (Fig. 4B) ranging from 1.52 nM to 10000.00 nM and DMSO vehicle control. Combination treatment with Compound 1 and Compound 2 (Fig. 4C).
[00286] Summary: Compound 2 induces cytostatic (growth-inhibitory) effect but not cell death, as noted by the dose-response curve remaining above the X-axis. Compound 1 induces cell-death (cytotoxic) effect (noted by the curve crossing the X-axis) at 1321.30 nM. Combination of Compound 1 and Compound 2 induces cell death effect at a lower dose (243.78 nM) than treatment with Compound 1 alone and of larger magnitude (88.3 compared to 65.3 percent cell death induced by Compound 1 alone). Effects of Compound 1 and Compound 2 Used Alone or in Combination with Each Other on Viability of NCI-H460 Lung Cancer Cell Line (Figure 5).
[00287] Figure 5. NCI-H460 cells were treated with a 9-point dose dilution of Compound 1 (Fig. 5A) or Compound 2 (Fig. 5B) ranging from 1.52 nM to 10000.00 nM and DMSO vehicle control. Combination treatment with Compound 1 and Compound 2 (Fig. 5C).
[00288] Summary: Compound 2 induces cytostatic (growth-inhibitory) effect but not cell death, as noted by the dose-response curve remaining above the X-axis. Compound 1 induces cell-death (cytotoxic) effect (noted by the curve crossing the X-axis) at 6839.12 nM. Combination of Compound 1 and Compound 2 induces cell death effect at a lower dose (1857.8 nM) than treatment with Compound 1 alone and of larger magnitude (66.3 compared to 18.0 percent cell death induced by Compound 1 alone). Effects of Compound 1 and Compound 2 Used Alone or in Combination with Each Other on Viability of NCI-H522 Lung Cancer Cell Line (Figure 6).
[00289] Figure 6. NCI-H522 cells were treated with a 9-point dose dilution of Compound 1 (Fig. 6A) or Compound 2 (Fig. 6B) ranging from 1.52 nM to 10000.00 nM and DMSO vehicle control. Combination treatment with Compound 1 and Compound 2 (Fig. 6C).
[00290] Summary: Compound 2 induces cytostatic (growth-inhibitory) effect but not cell death, as noted by the dose-response curve remaining above the X-axis. Compound 1 induces cell-death (cytotoxic) effect (noted by the curve crossing the X-axis) at 2338.84 nM. Combination of Compound 1 and Compound 2 induces cell death effect at a lower dose (625.17 nM) than treatment with Compound 1 alone and of larger magnitude (88.2 compared to 55.7 percent cell death induced by Compound 1 alone ). Effects of Compound 1 and Compound 2 Used Alone or in Combination with Each Other on Viability of NCI-H1755 Lung Cancer Cell Line (Figure 7).
[00291] Figure 7. NCI-H1755 cells were treated with a 9-point dose dilution of Compound 1 (Fig. 7A) or Compound 2 (Fig. 7B) ranging from 1.52 nM to 10000.00 nM and DMSO vehicle control. Combination treatment with Compound 1 and Compound 2 (Fig. 7C).
[00292] Summary: Compound 2 induces cytostatic (growth-inhibitory) effect but not cell death, as noted by the dose-response curve remaining above the X-axis. Compound 1 induces cell-death (cytotoxic) effect (noted by the curve crossing the X-axis) at 722.77 nM. Combination of Compound 1 and Compound 2 induces cell death effect at a lower dose (295.12 nM) than treatment with Compound 1 alone (370.37 nM). Effects of Compound 1 and Compound 2 Used Alone or in Combination with Each Other on Viability of Mia PaCa Pancreatic Cancer Cell Line (Figure 21).
[00293] This experiment is a replicate of the above-described assay on Mia PaCa cells. Figure 21. Mia PaCa Pancreatic Cancer Cell Line: Mia PaCa cells were treated with a 9-point dose dilution of Compound 1, or Compound 2, or both, ranging from 1.52 nM to 10000.00 nM, and DMSO vehicle control. Fig. 21A: dose response to Compound 1 alone (trend line with circles) or Compound 2 alone (trend line with triangles); Fig. 21B: combination treatment with Compound 1 and Compound 2 (trend line with circles) and untreated (trendline with triangles).
[00294] Summary: Compound 2 alone induces cytostatic (growth-inhibitory) effect but not cell death, as noted by the dose-response curve remaining above the X-axis. Compound 1 alone induces cell-death (cytotoxic) effect (noted by the curve crossing the X-axis) at 1238.80 nM. Combination of Compound 1 and Compound 2 induces cell death effect at a lower dose (533.33 nM) than treatment with Compound 1 alone and of larger magnitude (92.1 compared to 66.5 percent cell death induced by Compound 1 alone ). Effects of Compound 1 and Compound 2 Used Alone or in Combination with Each Other on Viability of Mia PaCa Pancreatic Cancer Cell Line (Figure 22).
[00295] This experiment is a replicate of the above-described assay on Mia PaCa cells. Figure 22. Mia PaCa Pancreatic Cancer Cell Line: Mia PaCa cells were treated with a 9-point dose dilution of Compound 1, or Compound 2, or both, ranging from 1.52 nM to 10000.00 nM, and DMSO vehicle control. Fig. 22A: dose response to Compound 1 alone (trend line with circles) or Compound 2 alone (trend line with triangles); Fig. 22B: combination treatment with Compound 1 and Compound 2 (trend line with circles).
[00296] Summary: Compound 2 induces cytostatic (growth-inhibitory) effect but not cell death, as noted by the dose-response curve remaining above the X-axis. Compound 1 induces cell-death (cytotoxic) effect (noted by the curve crossing the X-axis) at 1534.61 nM. Combination of Compound 1 and Compound 2 induces cell death effect at a lower dose (628.06 nM) than treatment with Compound 1 alone and of larger magnitude (88.9 compared to 49.9 percent cell death induced by Compound 1 alone ). Effects of Compound 1 and Compound 2 Used Alone or in Combination with Each Other on Viability of HS294T Melanoma Cancer Cell Line (Figure 23).
[00297] This experiment is a replicate of the above-described assay on HS294T melanoma cells. Figure 23. HS294T MelanomaCancer Cell Line: HS294T cells were treated with a 9-point dose dilution of Compound 1, or Compound 2, or both, ranging from 1.52 nM to 10000.00 nM, and DMSO vehicle control. Fig. 23A: dose response to Compound 1 alone (trend line with circles) or Compound 2 alone (trend line with triangles); Fig. 23B: combination treatment with Compound 1 and Compound 2 (trend line with circles). [00298] Summary: Compound 2 alone induces cytostatic (growth-inhibitory) effect but not cell death, as noted by the dose-response curve remaining above the X-axis. Compound 1 alone induces cell-death (cytotoxic) effect (noted by the curve crossing the X-axis) at 1202.26 nM. Combination of Compound 1 and Compound 2 induces cell death effect at a lower dose (276.0 nM) than treatment with Compound 1 alone and of larger magnitude (95.6 compared to 86.9 percent cell death induced by Compound 1 alone).
[00299] As these data demonstrate, relative to the doses of Compound 1 or Compound 2 resulting in a cell killing effect from treatment with a single agent, combining Compound 1 with Compound 2 at lower doses of each inhibitor was found to induce the same or better cell killing effect as Compound 1 or Compound 2 alone at the higher doses. [00300] Combining these inhibitors may allow them to be used at doses lower than would be - necesary to achieve tumor cell killing if administered alone and thus may be beneficial in achieving the desired tumor cell death effect while mitigating possible adverse effects such as toxicity liabilities of either drug alone administered at higher doses. Example 3
Combination Experiments with Titration of Compound 1 and Fixed Doses of Compound 2:
Maximum Effect Study
[00301] Data obtained from the assay described below in Example 6 can be used to determine the maximum effect (Emax) of Compound 1 in combination with Compound 2. Emax is the effect a compound has on cell growth at a given concentration It is evaluated both relative to control treatment and relative to cell number at time zero, and usually ranges from 100% to - 100%. By way of example, an Emax of 100% indicates that the treated cells have grown as fast as the control cells; an Emax of 20% indicates that the treated cells have grown slower and their growth rate was 20% of the growth rate of cells in control treated wells; an Emax of -20% indicates that the cells have declined in number compared to the number of cells seeded at day zero (i.e., by 20%); an Emax of -100% indicates that the cells have declined in number compared to the number of cells seeded at day zero (i.e., by 100%, that is, all cells are dead). Values of >100% indicate that the cells in the treated well have grown faster than the cells in the control well, typically indicating no negative effect (as opposed to a positive effect).
[00302] As depicted in Figure 8, a moderate enhancement in Emax is seen when combining Compound 1 with Compound 2, a TOR inhibitor. Compound 1 does not kill any of the tested lines at 1 μM, and in combination with Compound 2, cell death is observed in 1/6 tested lines, NCI-H727. Emax in this line reached -16% when Compound 1 was combined with 1μM Compound 2.
Example 4
Calcusyn Analysis and Colony Forming Assay [00303] Calcusyn Analysis: The following assays were conducted with Compound 1 and Compound 2 and demonstrate that in certain cell lines synergy is observed upon treatment with a combination of Compound 1 and Compound 2. The Calcusyn assay was carried out over 5 days. On day 1, cells were plated into 96 well plates, in 100 μL media (cell number calculated to give 50-70% fluency on day 5). On day 2, drug dilutions were prepared at 2x final concentration in media, 100 L /well were added, and 4-5 replicates/drug concentration were completed. On day 5, media was removed from the plate. 100 μL were added of 10% TCA/well and fixed for 1 hour at 4 °C. The plates were washed three times in tap water and allowed to dry. The plates were then stained with 0.2% SRB stain for thirty minutes at room temperature. Plates were then washed three times in 1% acetic acid and allowed to dry. Dye was resuspended in 100 μL /well of 10 mM Tris and the plate was read at 565 nM wavelength. Calcusyn analysis of the data gave a combination index value (CI-value) which offers a quantitative definition for additive effect (CI = 1), synergism (CI < 1), and antagonism (CI > 1) of the tested drug combination. This combination was tested for synergy at both ED50 (i.e., the dose concentration determined to produced quantal effect in 50% of cells) and ED75 (i.e., the dose concentration determined to produced quantal effect in 75% of cells). The following cell lines were tested: B-raf mutant vemurafenib-resistant melanoma cell line 1 (BRAF status: V600E(homo)); B-raf mutant vemurafenib-resistant melanoma cell line 2 (BRAF status: V600E(het)); B-raf mutant vemurafenib-resistant melanoma cell line 3 (BRAF status: V600E(homo)); B-raf mutant vemurafenib-resistant melanoma cell line 4 (BRAF status: V600E(het)); B-raf mutant vemurafenib-resistant melanoma cell 5 (BRAF status: V600 E(het)); B-raf mutant vemurafenib- resistant melanoma cell line 6 (BRAF status: V600E(het)); B-raf mutant vemurafenib-resistant melanoma cell line 7 (BRAF status: V600E); B-raf mutant vemurafenib-resistant melanoma cell line 8 (BRAF status: V600E).
[00304] As depicted in Figure 9, a Calcusyn analysis indicated that Compound 1 and Compound 2 were generally synergistic across a panel of B-raf mutant vemurafenib resistant melanoma cell lines (as evidenced by a CI-value of <1). This combination was tested for synergy at both ED50 (i.e., the dose concentration determined to produced quantal effect in 50% of cells) and ED75 (i.e., the dose concentration determined to produced quantal effect in 75% of cells). [00305] Colony Assays: Compound 1 (free base) with Compound 2 were also tested for in colony assays. Cells were seeded at low density in 6-well plates. Cells were treated for two to four weeks with Compound 1 and/or Compound 2, as either single agents or in combination. Colonies were fixed and stained with 0.1% crystal violet solution and then counted.
[00306] Colony assays were performed to determine the ability of a B-raf mutant vemurafenib-resistant melanoma cell line (i.e., cell line 8) to generate colonies after treatment with a combination of Compounds 1 and 2. Figure 10 contains a representative graph depicting the results of these assays.
[00307] Colony assays were performed to determine the ability of a B-raf mutant vemurafenib-resistant melanoma cell line (i.e., cell line 3) to generate colonies after treatment with a combination of Compounds 1 and 2. Figure 11 contains a representative graph depicting the results of these assays.
[00308] Combination treatement with Compound 1 and Compound 2 was also assessed in 4 NRAS mutant melanoma cell lines A-D. As depicted in Figure 12, the combination of Compound 1 and Compound 2 showed CI-values of <1 displaying synergy against NRAS mutant melanoma cell lines A-D. Example 5
KRAS G12D cell line CRC PDX model
[00309] A combination PD study of Compound 1 (in the form of the phosphate salt) and Compound 6 was run in a KRAS G12D cell line CRC (i.e., colorectal cancer) PDX model. Briefly, the study design is as described below. Greater than 60 athymic nude mice were implanted with KRAS G12D CRC patient-derived xenographs. The drugs were formulated in 5% Captisol and 0.4% Tween80. The mice were treated Monday through Friday for 28 days, or until tumors reached 1250 mm3. Plasma was isolated on day 28 at 0.5, 1, 2, 4, 8 and 24 hours. The tumors were harvested on day 28 with 1/3 of the tumor formalin fixed and the remaining 2/3 of the tumor snap frozen.
[00310] As depicted in Figure 13, the combination of Compound 1 (phosphate salt) and Compound 6 showed an decrease in tumor volume over treatment with either Compound 1 (phosphate salt) or Compound 6 alone. Single agent Compound 6 (15 mg/kg) reduced tumor volume 7%. Single agent Compound 1 (50 mg/kg) reduced tumor volume 20%. Single agent Compound 1 (100 mg/kg) reduced tumor volume 30%. Combination treatements reduced tumor volume 34%-41%. Example 6
Combination Experiments with Titration of Compound 1 and Fixed Doses of Compound 2:
Synergy Study
[00311] The cells were plated at density 3000 cells/well in 90 µL of growth media on 96 well clear bottom black-well plates (Corning Cat# 3904) and incubated overnight under standard cell culture growth conditions at 37 0C, 5% CO2. The outer most rows and columns of wells were filled with culture media, without cells, to avoid evaporation effects on subsequent readouts.
[00312] The following day, one plate for each cell line was used for“Day 0” cell growth control readout, and cell viability was measured with CellTiter Glo (Promega) reagent according to manufacturer specifications. Briefly, Day 0 control plates were equilibrated to room temperature for 30 minutes in a tissue culture hood, followed by addition of 90 µL of CellTiter Glo reagent (a volume equal to cell media volume) to each well. The plates were covered with foil to protect from light and placed on shaker at low speed for 5 minutes followed by 10 minutes incubation without shaking. CellTiter Glo reagent signal was read on a spectramax L luminescence detector and data were processed using Excel and Prism software.
[00313] At the same time (while Day 0 plate was equilibrate to room temperature) cells in the remaining plates were treated with Compound 1 (free base), Compound 2, (i.e., a TOR inhibitor), or a combination of Compound 1 and Compound 2, or DMSO vehicle control, as specified for each cell line. For cell treatment compounds were diluted from starting concentration 10 mM stock in DMSO to 10 µM starting in-well concentration (3 dilution steps first serially in DMSO, then in cell growth media), such that resulting DMSO concentration in the growth media in wells containing cells was 0.1%. The cells were treated with 9 point 3-fold dilutions of Compound 1 and a constant concentration of Compound 2, or DMSO vehicle control. Thus, the final concentrations of Compound 1 in treatment wells were: (in nM) 10000.00, 3333.33, 1111.11, 370.37, 123.46, 41.15, 13.72, 4.57, 1.52, and 0. Compound 1 was combined at each dilution level with a selected constant dose of Compound 2. Each treatment was contained in 10 µL treatment media added to the respective wells. Each concentration was tested in triplicate.
[00314] Seventy two hours later“Day 3” cell viability was measured by adding 100 µL CellTiter Glo reagent to each of the treatment wells, equal to the volume of media and compound. Cell viability was assessed as described for“Day 0” control.
[00315] Interpretation of Synergism Plots (“Volcano Plots”): Figures 14-17, 19-20, and 24-29 depict volcano plots generated using data generated in the manner described above in order to detect synergy for combination drug treatment of certain cell lines. Assays were completed as described above using Compound 1 and Compound 2. The x-axis shows the 9 point dose titration of Compound 1 and DMSO control; the y-axis shows three individual fixed concentrations of Compound 2 in each experiment; and the z-axis shows the magnitude of the effect, with different patterns denoting different percent improvement over the additive effect of the specific compound combinations, as specified in each figure. Where a significant percent improvement over the additive effect is observed (e.g., >10%), the combination is said to be synergistic. Accordingly, in instances in which a volcano plot peak reaches a height corresponding to a >10% increase beyond additive effect, that plot is described as synergistic, meaning synergy over 10%.
[00316] As used herein, synergy is defined as achieving a greater than additive effect with a particular drug combination. An additive effect is a predicted additive effect, assuming independent action of each individual drug (i.e., Bliss independence). If a particular combination of drugs affords results which exceed the predicted additive effect, that combination is considered to be synergistic. If a particular combination of drugs affords results which fall short of the predicted additive effect, that combination is considered to be antagonistic.
[00317] By way of example, if each of two drugs are known to independently inhibit 50% of a particular substrate population, the additive effect is determined by assuming that the first drug will inhibit 50% of the substrate population and the second drug will inhibit 50% of the remaining substrate population, leading to a predicted additive effect of 75% inhibition. In such a case, inhibition of greater than 75% of a substrate population with combination treatment using the two drugs suggests synergy. Likewise, in such a case, inhibition of less than 75% of a substrate population with combination treatment using the two drugs suggests antagonism. [00318] Figures 25A-B, 26A-B, 27A-D, 28A-B, and 29A-B-29, described below, depict representative volcano plots for combination treatment of Compound 1 and Compound 2 on various cell lines.
[00319] Figure 14 depicts a volcano plot at section (a) generated using data obtained for HCT-116 colorectal cancer cells treated with a combination of Compound 1 and Compound 2 at concentrations of 0.1 M, 0.3 μM and 1.0 μM. The height of the plot peak suggests a synergistic effect is achieved at certain combination doses. Relevant dose response assay data are provided at section (b) of Figure 14.
[00320] Figure 15 depicts a volcano plot at section (a) generated using data obtained for HCT-116 colorectal cancer cells treated with a combination of Compound 1 and Compound 2 at concentrations of 0.03 μM, 0.3 μM and 1.0 μM. The height of the plot peak suggests a synergistic effect is achieved at certain combination doses. Relevant dose response assay data are provided at section (b) of Figure 15.
[00321] Figure 16 depicts a volcano plot at section (a) generated using data obtained from a replicate experiment for HCT-116 colorectal cancer cells treated with a combination of Compound 1 and Compound 2 at concentrations of 0.03 μM, 0.3 μM and 1.0 μM. The height of the plot peak suggests a synergistic effect is achieved at certain combination doses. Relevant dose response assay data are provided at section (b) of Figure 16.
[00322] Figure 17 depicts a volcano plot at section (a) generated using data obtained for MiaPaCa pancreatic cancer cells treated with a combination of Compound 1 and Compound 2 at concentrations of 0.1 μM, 0.3 μM and 1.0 μM. The height of the plot peak suggests an additive effect is achieved at certain combination doses. Relevant dose response assay data are provided at section (b) of Figure 17.
[00323] Figure 19 depicts a volcano plot at section (a) generated using data obtained for MiaPaCa pancreatic cancer cells treated with a combination of Compound 1 and Compound 2 at concentrations of 0.03 μM, 0.3 μM and 1.0 μM. The height of the plot peak suggests a an additive effect is achieved at certain combination doses. Relevant dose response assay data are provided at section (b) of Figure 19.
[00324] Figure 20 depicts a volcano plot at section (a) generated using data obtained from a replicate experiment for MiaPaCa pancreatic cancer cells treated with a combination of Compound 1 and Compound 2 at concentrations of 0.03 μM, 0.3 μM and 1.0 μM. The height of the plot peak suggests additive effect is achieved at certain combination doses. Relevant dose response assay data are provided at section (b) of Figure 20.
[00325] Figure 24 depicts a volcano plot generated using data obtained for Calu-1 lung cancer cells with a combination of Compound 1 and Compound 2 at concentrations of 0.1 μM, 0.3 μM and 1.0 μM. This plot suggests that additive effects are observed for this combination at certain dosages.

Claims

CLAIMS We claim:
1. A method of treating, stabilizing, or lessening the severity or progression of one or more diseases or disorders associated with one or both of ERK1 and ERK2, comprising administering to a patient in need thereof a composition comprising Compound 1:
Figure imgf000087_0001
or a pharmaceutically acceptable salt thereof, in combination with a TOR inhibitor.
2. The method according to claim 1, wherein the TOR inhibitor is an mTOR inhibitor.
3. The method according to claim 1, wherein the TOR inhibitor is an allosteric mTOR inhibitor.
4. The method according to claim 1, wherein Compound 1 and the TOR inhibitor are administered simultaneously.
5. The method according to claim 1, wherein Compound 1 and the TOR inhibitor are administered sequentially.
6. The method according to claim 1, wherein the TOR inhibitor is selected from rapamycin, AP23573 (Deforolimus), AP-23675, AP-23841, ABT-578 (Zotarolimus), CCI779 (Temsirolimus), RAD-001 (Everolimus), 7-epi-rapamycin, 7-thiomethyl-rapamycin, 7-epi- trimethoxyphenyl-rapamycin, 7-epi-thiomethyl-rapamycin, and 7-demethoxy-rapamycin, 32- demethoxy-rapamycin, 2-desmethyl-rapamycin, 42-O-(2-hydroxy)ethyl rapamycin, Ridaforolimus, AZD8055, OSI-027, INK-128 (MLN0128), wortmannin, and LY29004.
7. The method according to claim 1, wherein the TOR inhibitor is a compound of Formula (I):
Figure imgf000088_0001
and pharmaceutically acceptable salts, clathrates, solvates, stereoisomers, tautomers, metabolites, isotopologues and prodrugs thereof, wherein:
R1 is substituted or unsubstituted C1-8 alkyl, substituted or unsubstituted aryl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocyclyl, or substituted or unsubstituted heterocyclylalkyl;
R2 is H, substituted or unsubstituted C1-8 alkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocyclyl, substituted or unsubstituted heterocyclylalkyl, substituted or unsubstituted aralkyl, or substituted or unsubstituted cycloalkylalkyl; and
R3 is H, or a substituted or unsubstituted C1-8 alkyl.
8. The method according to claim 7, wherein the TOR inhibitor is selected from any of Compound 2, Compound 3, and Compound 4, or a pharmaceutically acceptable salt thereof.
9. The method according to claim 1, wherein the TOR inhibitor is selected from those described in the publications listed in either of Table A or Table B.
10. The method according to claim 1, wherein the TOR inhibitor is selected from a rapamycin.
11. The method according to claim 1, wherein the TOR inhibitor is selected from Everolimus, Temsirolimus, Ridaforolimus, AZD8055, OSI-027, and INK-128.
12. The method according to any one of claims 1-11, wherein the disease or condition is a cancer.
13. The method according to claim 12, wherein the disease or condition is a MAPK pathway- mediated cancer.
14. The method according to claim 12, wherein the cancer is a BRAF-mutated cancer.
15. The method according to claim 14, wherein the BRAF-mutated cancer is a BRAFV600 mutated cancer.
16. The method according to claim 15, wherein the BRAFV600 mutated cancer is selected from BRAFV600E, BRAFV600K, BRAFV600R, or BRAFV600D.
17. The method according to claim 12, wherein the cancer is a RAS-mutated cancer.
18. The method according to claim 17, wherein the RAS-mutated cancer is a KRAS-mutated cancer.
19. The method according to claim 18, wherein the KRAS-mutated cancer is selected from KRASG12C/D/V, KRASG13C/D, and KRASQ61L/H/R
20. The method according to claim 17, wherein the RAS-mutated cancer is an NRAS- mutated cancer.
21. The method according to claim 20, wherein the NRAS-mutated cancer is selected from NRASQ61R, NRASQ61K, NRASQ61L, and NRASQ61H.
22. The method according to claim 17, wherein the RAS-mutated cancer is an HRAS- mutated cancer.
23. The method according to claim 22, wherein the HRAS-mutated cancer is selected from HRASG12V, HRASQ61R, and HRASG12S.
 
24. The method according to claim 12, wherein the cancer is selected from breast, colorectal, endometrial, hematological, leukemia (e.g., AML), liver, lung, melanoma, ovarian, pancreatic, prostate, and thyroid.
25. The method according to claim 24, wherein the cancer is selected from breast, colorectal, endometrial, liver, lung, melanoma, ovarian, pancreatic, and thyroid.
26. The method according to claim 25, wherein the cancer is selected from colorectal, lung, melanoma, and pancreatic
27. The method according to claim 26, wherein the cancer is selected from colorectal, melanoma, and pancreatic.
28. The method according to claim 27, wherein the cancer is melanoma.
29. The method according to claim 28, wherein the melanoma is Vemurfenib-resistant, dabrafenib-resistant, or encorafenib-resistant BRAF-mutated melanoma.
30. The method according to claim 28, wherein the melanoma is NRAS-mutated melanoma.
31. The method according to claim 28, wherein the melanoma is uveal melanoma.
32. The method according to claim 27, wherein the cancer is colorectal cancer.
33. The method according to claim 32, wherein the colorectal cancer is BRAF-mutated colorectal cancer.
34. The method according to claim 32, wherein the colorectal cancer is KRAS-mutated colorectal cancer.
35. The method according to claim 27, wherein the cancer is pancreatic cancer.
36. The method according to claim 35, wherein the cancer is KRAS-mutated pancreatic cancer.
37. A system for treating, stabilizing or lessening the severity of one or more diseases or conditions associated with one or more of ERK1 and ERK2, the system comprising Compound 1:
Figure imgf000091_0001
or a pharmaceutically acceptable salt ation with a TOR inhibitor.
38. The system according to claim 37, wherein the TOR inhibitor is selected from rapamycin, AP23573 (Deforolimus), AP-23675, AP-23841, ABT-578 (Zotarolimus), CCI779 (Temsirolimus), RAD-001 (Everolimus), 7-epi-rapamycin, 7-thiomethyl-rapamycin, 7-epi- trimethoxyphenyl-rapamycin, 7-epi-thiomethyl-rapamycin, and 7-demethoxy-rapamycin, 32- demethoxy-rapamycin, 2-desmethyl-rapamycin, 42-O-(2-hydroxy)ethyl rapamycin, Ridaforolimus, AZD8055, OSI-027, INK-128, MLN0128, wortmannin, and LY29004.
39. The system according to claim 37, wherein the TOR inhibitor is a compound of Formula (I)
Figure imgf000092_0001
and pharmaceutically acceptable salts, clathrates, solvates, stereoisomers, tautomers, metabolites, isotopologues and prodrugs thereof, wherein:
R1 is substituted or unsubstituted C1-8 alkyl, substituted or unsubstituted aryl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocyclyl, or substituted or unsubstituted heterocyclylalkyl;
R2 is H, substituted or unsubstituted C1-8 alkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocyclyl, substituted or unsubstituted heterocyclylalkyl, substituted or unsubstituted aralkyl, or substituted or unsubstituted cycloalkylalkyl; and
R3 is H, or a substituted or unsubstituted C1-8 alkyl.
40. The system according to claim 39, wherein the TOR inhibitor is selected from any of Compound 2, Compound 3, and Compound 4, or a pharmaceutically acceptable salt thereof.
41. A composition comprising Compound 1, or a pharmaceutically acceptable salt thereof, a TOR inhibitor, and a pharmaceutically acceptable carrier, adjuvant, or vehicle.
PCT/US2015/044931 2014-08-13 2015-08-12 Combinations of an erk inhibitor and a tor inhibitor and related methods WO2016025651A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201462037067P 2014-08-13 2014-08-13
US62/037,067 2014-08-13

Publications (1)

Publication Number Publication Date
WO2016025651A1 true WO2016025651A1 (en) 2016-02-18

Family

ID=55304600

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2015/044931 WO2016025651A1 (en) 2014-08-13 2015-08-12 Combinations of an erk inhibitor and a tor inhibitor and related methods

Country Status (1)

Country Link
WO (1) WO2016025651A1 (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN106508907A (en) * 2016-10-21 2017-03-22 全南厚朴生态林业有限公司 Chimonanthus grammatus rooting agent formula and using method
CN111436337A (en) * 2020-03-04 2020-07-24 沈阳农业大学 Application of rapamycin target protein activator MHY1485 in promotion of tomato seed germination and seedling growth and development
US11040027B2 (en) 2017-01-17 2021-06-22 Heparegenix Gmbh Protein kinase inhibitors for promoting liver regeneration or reducing or preventing hepatocyte death

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100029610A1 (en) * 2008-06-27 2010-02-04 Avila Therapeutics, Inc. Heteroaryl Compounds and Uses Thereof
WO2012170976A2 (en) * 2011-06-10 2012-12-13 Merck Patent Gmbh Compositions and methods for the production of pyrimidine and pyridine compounds with btk inhibitory activity

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100029610A1 (en) * 2008-06-27 2010-02-04 Avila Therapeutics, Inc. Heteroaryl Compounds and Uses Thereof
WO2012170976A2 (en) * 2011-06-10 2012-12-13 Merck Patent Gmbh Compositions and methods for the production of pyrimidine and pyridine compounds with btk inhibitory activity

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
LIU, Q ET AL.: "Developing Irreversible Inhibitors of the Protein Kinase Cysteinome", CHEMISTRY & BIOLOGY, vol. 20, 2013, pages 146 - 159 *
WALTER, AO ET AL.: "Discovery of a Mutant-Selective Covalent Inhibitor of EGFR that Overcomes T790M Mediated Resistance in NSCLC", CANCER DISCOVERY, vol. 3, no. 12, 2013, pages 1405 - 1415, XP055230789, DOI: doi:10.1158/2159-8290.CD-13-0314 *
ZHOU, W ET AL.: "Novel mutant-selective EGFR kinase inhibitors against EGFR T790M", NATURE, vol. 462, no. 7276, 2009, pages 1070 - 1074, XP055053374, DOI: doi:10.1038/nature08622 *

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN106508907A (en) * 2016-10-21 2017-03-22 全南厚朴生态林业有限公司 Chimonanthus grammatus rooting agent formula and using method
US11040027B2 (en) 2017-01-17 2021-06-22 Heparegenix Gmbh Protein kinase inhibitors for promoting liver regeneration or reducing or preventing hepatocyte death
CN111436337A (en) * 2020-03-04 2020-07-24 沈阳农业大学 Application of rapamycin target protein activator MHY1485 in promotion of tomato seed germination and seedling growth and development
CN111436337B (en) * 2020-03-04 2021-10-22 沈阳农业大学 Application of rapamycin target protein activator MHY1485 in promotion of tomato seed germination and seedling growth and development

Similar Documents

Publication Publication Date Title
CA2788678C (en) Identification of lkb1 mutation as a predictive biomarker for sensitivity to tor kinase inhibitors
US9375443B2 (en) Method for treating advanced non-small cell lung cancer (NSCLC) by administering a combination of a TOR kinase inhibitor and azacitidine or erlotinib
AU2015213353B2 (en) Treatment of cancer with TOR kinase inhibitors
AU2013203153B2 (en) Treatment of cancer with TOR kinase inhibitors
US10391092B2 (en) Methods for treating cancer using dihydropyrazino-pyrazine compound combination therapy
AU2014254052B2 (en) Treatment of cancer with dihydropyrazino-pyrazines
JP2018065820A (en) Treatment of cancer with tor kinase inhibitors
WO2016025651A1 (en) Combinations of an erk inhibitor and a tor inhibitor and related methods
AU2015213400B2 (en) Treatment of cancer with TOR kinase inhibitors
AU2015201138B2 (en) Treatment of cancer with TOR kinase inhibitors

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: 15831698

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: 15831698

Country of ref document: EP

Kind code of ref document: A1